Next Article in Journal
Circulating Tumor DNA—A Novel Biomarker of Tumor Progression and Its Favorable Detection Techniques
Previous Article in Journal
Prognostic Significance of p16 and Its Relationship with Human Papillomavirus Status in Patients with Penile Squamous Cell Carcinoma: Results of 5 Years Follow-Up
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Status of Novel Agents for the Treatment of B Cell Malignancies: What’s Coming Next?

Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(24), 6026; https://doi.org/10.3390/cancers14246026
Submission received: 7 November 2022 / Revised: 29 November 2022 / Accepted: 3 December 2022 / Published: 7 December 2022
(This article belongs to the Section Cancer Biomarkers)

Abstract

:

Simple Summary

Since the identification of a large variety of biomarkers in B cell malignancies as being driving factors for tumor progression and patient prognosis, their targeting may confer valuable options for the treatment of these diseases. Over the past 20 years, the permanent development of a multitude of inhibitors acting on survival-associated biomarkers has made it into clinical evaluation in B lymphoid tumors. Although certain drugs approved by the US Food and Drug Administration improve clinical outcome, some patients do not respond and others relapse. This review summarizes the current state-of-the-art, provides a summary of new, safer, more selective inhibitors currently under evaluation in clinical trials, and highlights the emerging positioning of metabolic drugs in tumor B cell biology as a promising strategy to be translated into clinical practice.

Abstract

Resistance to death is one of the hallmarks of human B cell malignancies and often contributes to the lack of a lasting response to today’s commonly used treatments. Drug discovery approaches designed to activate the death machinery have generated a large number of inhibitors of anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 family and the B-cell receptor (BCR) signaling pathway. Orally administered small-molecule inhibitors of Bcl-2 protein and BCR partners (e.g., Bruton’s tyrosine kinase and phosphatidylinositol-3 kinase) have already been included (as monotherapies or combination therapies) in the standard of care for selected B cell malignancies. Agonistic monoclonal antibodies and their derivatives (antibody–drug conjugates, antibody–radioisotope conjugates, bispecific T cell engagers, and chimeric antigen receptor-modified T cells) targeting tumor-associated antigens (TAAs, such as CD19, CD20, CD22, and CD38) are indicated for treatment (as monotherapies or combination therapies) of patients with B cell tumors. However, given that some patients are either refractory to current therapies or relapse after treatment, novel therapeutic strategies are needed. Here, we review current strategies for managing B cell malignancies, with a focus on the ongoing clinical development of more effective, selective drugs targeting these molecules, as well as other TAAs and signaling proteins. The observed impact of metabolic reprogramming on B cell pathophysiology highlights the promise of targeting metabolic checkpoints in the treatment of these disorders.

1. Background and Introduction

The balance between cell death and cell proliferation contributes to the maintenance of tissue homeostasis. Cell death evasion is a hallmark of human cancers (including B cell malignancies; Table 1) and might contribute to treatment resistance [1]. A number of molecular pathways underpin cell growth and cell survival in B cell malignancies (Figure 1). For example, anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 (BCL2) family (such as Mcl-1, Bcl-2, Bcl-xL, and Bcl-w) are overexpressed in most B cell malignancies (Table 1) as a result of genetic lesions or changes in signal transduction [2,3]. This expression is associated with a poor prognosis [3,4,5,6]. Anti-apoptotic BCL2 proteins interact with the pro-apoptotic “BH3-only” proteins (such as Bim, Bid, Puma, Bad, and Noxa) or bind and sequester the apoptotic effectors Bax and Bak in an inactive form (Figure 1a) [3,7]. The pro-apoptotic proteins induce apoptosis by activating Bax and Bak either directly or indirectly via inhibition of the anti-apoptotic proteins (Figure 1a) [3,7]. Active B-cell receptor (BCR) signaling also contributes to cell survival, proliferation, and resistance to apoptosis in B cell malignancies [8,9,10,11,12] (Table 1). BCR-mediated proliferation of malignant B cells may be caused by BCR pathway mutations and/or chronic active stimulation of the BCR [8,9,10,11,12]. The mechanism of BCR pathway activation in B cell malignancies is complex and has been well reviewed by Burger and Wiestner [13]; it includes continuous BCR stimulation by microbial antigens or autoantigens, activating mutations within the BCR complex or downstream signaling partners, and antigen-independent BCR signaling [13]. The BCR pathway includes a large array of kinases, adaptor proteins and transcription factors (such as NF-κB); in particular, Bruton tyrosine kinase (BTK) and phosphatidylinositol-3 kinase (PI3K) are critical signaling enzymes involved in uncontrolled B cell proliferation and survival [10,11,12,13] (Figure 1b). Lastly, high levels of expression of certain tumor-associated antigens (TAAs; CD19, CD20, CD22, and CD38) in most B cell malignancies are correlated with a high proliferation rate and disease progression (Table 1) [14,15,16,17]. These TAAs are physically associated with the BCR [18,19,20,21] and are involved in modulating BCR-dependent (Figure 1b) and BCR-independent proliferation/survival signals (Figure 1c) [13,15,16,17,18,20,21,22,23,24,25,26].
Over the last 25 years, greater knowledge of the detrimental roles of these survival-associated proteins in malignant B cell pathologies have prompted the development of a large array of inhibitors. These novel anticancer agents include small molecules that target BCL2 members, BTK, or PI3K, and mAbs and their derivatives (mono/bispecific Ab, antibody–drug conjugate/ADC, antibody–radioisotope conjugate/ARC, bispecific T cell engager/BiTE, and chimeric antigen receptor-modified T cells/CAR-T) against TAAs. The use of some of these novel agents have been authorized by the US Food and Drug Administration (FDA) [27]. Although these drugs improve clinical outcomes when used alone or in combination with other treatments, some patients do not respond, and others relapse after an initial response [28,29,30,31,32,33,34]. As such, efforts to develop more effective, selective inhibitors against the same targets or new targets in malignant B cells must be continued. Specifically, novel mAbs (acting on other TAAs) and small-molecule inhibitors (acting on survival-associated proteins, such as signaling proteins, proteases, immune checkpoints, etc.) are in preclinical or clinical development. Here, we first review the current management of B lymphoid tumors with FDA-approved drugs targeting the above-cited TAAs, Bcl-2, or BTK/PI3K. We then focus on the clinical efficacy of newly developed drug candidates for inhibiting current and novel protein targets and overcoming some forms of resistance. Lastly, we review perspectives for therapy, with emphasis on the importance of metabolic reprogramming in tumor B cell biology and opportunities for inhibiting this process.
Table 1. Characteristics of the main B cell malignancies.
Table 1. Characteristics of the main B cell malignancies.
B Cell MalignancyPathophysiologyExpression of BCL2 Prosurvival ProteinsActive (Pre-)BCR Signaling Surface Markers That Are Strongly ExpressedReferences
B-ALLTransformation and expansion of lymphatic B progenitor cellsBcl-2, Mcl-1, Bcl-xLpre +CD19 CD20
CD22 CXCR4
[7,35,36,37,38,39,40,41]
HCLAccumulation of CD5+ B cells most in the blood, bone marrow and spleenBcl-2 > Mcl-1+CD19 CD20
CD22 CD38
CXCR4
[2,11,40,42,43]
CLLAccumulation of CD5+ B cells primarily in the blood and bone marrowHigh Bcl-2 > Mcl-1 >> Bcl-xL+CD19 CD20
CD38 (poor prognosis)
BAFF-R ROR1
CXCR4
[3,7,36,38,40,41,44,45]
SLL
(B-NHL)
Accumulation of CD5+ B cells most in the lymph nodesBcl-2, Mcl-1,
Bcl-xL
+CD19 CD20
CD38 (poor prognosis) ROR1
[40,44,45,46,47]
MMAccumulation of clonal, Ig secreting plasma cells in the bone marrow High Bcl-2, Mcl-1, Bcl-xL, Bcl-w+CD19 CD22
CD38 BCMA
CD13 ADAM17
ROR1 CXCR4
[3,7,40,41,48,49,50,51]
FL
(B-NHL)
Extensive proliferation and accumulation of abnormal B cells in lymph nodesHigh Bcl-2, Bcl-w and Bcl-xL > Mcl-1−/+CD19 CD20
CD22 CD38
CXCR4
[3,7,40,41,45,48,52]
MCL
(B-NHL)
Development of abnormal B cells in the mantle zone of lymph nodes, spleen, bone marrow, blood, and gastrointestinal tractHigh Bcl-w and Bcl-2 > Mcl-1+CD19 CD20
CD22 CD38
ROR1 CXCR4
[3,7,40,41,47,52,53]
MZL
(B-NHL)
Development of abnormal B cells in the marginal zones of lymph tissueHigh Bcl-2 and Bcl-w > Bcl-xL+CD19 CD20
CD22 CXCR4
[3,10,40,41,52,54,55]
DLBCL
(B-NHL)
Development of abnormal B cells in germinal centers of secondary lymphoid organsBcl-2 and Bcl-w > Mcl-1, Bcl-xL+CD19 CD20
CD22 CD38
CXCR4
[3,7,36,40,41,45,52,56,57]
WM
(B-NHL)
Proliferation of clonal, IgM-secreting plasma cells in the lymph nodes and bone marrowHigh Bcl-2 > Bcl-xL and Mcl-1+CD19 CD20
CD38
CXCR4 (mutation)
[8,40,46,58,59]
ADAM17, a disintegrin and metalloprotease 17; B-ALL, B-acute lymphoblastic leukemia; BAFF-R, B-cell activating factor receptor; BCMA, B cell maturation antigen; BCR, B-cell receptor; B-NHL, B cell non-Hodgkin lymphoma; CLL, chronic lymphocytic leukemia; CXCR4, CXC chemokine receptor 4 specific for stromal cell-derived factor-1. DLBCL, diffuse large B cell lymphoma; FL, follicular lymphoma; HCL, hairy cell leukemia; MCL, mantle cell lymphoma; MM, multiple myeloma; MZL, marginal zone lymphoma; ROR1, receptor tyrosine kinase-like orphan receptor; SLL, small lymphocytic lymphoma; WM, Waldenström’s macroglobulinemia. Present on precursor B lymphocytes, pre-BCR differs from conventional BCRs in that it possesses a germ line-encoded surrogate light chain which is associated with the signal transduction machinery via heavy chain proteins that have been generated by productive rearrangement of the immunoglobulin HC genes.

2. Current Treatments and Clinical Trials of B Cell Malignancies Using Drugs That Target BCL2, BTK, PI3K Proteins, and TAAs (CD19, CD20, CD22, CD38)

The drugs currently approved by the FDA for the treatment of B cell malignancies [27] still include the first-in-class anti-CD20 agent (rituximab), the first-in-class Bcl-2 inhibitor (venetoclax), the first-in-class BTK inhibitor (ibrutinib), and the first-in-class PI3K inhibitor (idelalisib). Figure 2 charts these key discoveries, which led to the development of more efficacious treatments (Figure 2a) and new indications for clinical evaluation (Figure 2b). These agents include BCL2, BTK, and PI3K inhibitors, together with mAbs and their derivatives (ADC, ARC, bispecific Ab, and CAR-T).

2.1. BCL2 Inhibitors

Over the last 20 years, significant efforts have been made to develop the therapeutic potential of agents targeting BCL2 family members [3,30,60]. The broad panel of small molecules developed to mimic the action of anti-apoptotic proteins includes navitoclax/ABT-263 (targeting Bcl-xL, Bcl-2, and Bcl-w), obatoclax/GX15-070 (targeting Bcl-2, Bcl-xL, Mcl-1, and Bcl-w), and venetoclax/ABT-199 (targeting Bcl-2 only); all have entered clinical trials [3,61,62]. Navitoclax and obatoclax induced severe adverse events, and the trials were stopped [3,61,62]. Venetoclax was approved in 2016 for treatment of previously treated CLL patients with a 17p deletion [3,60]. Resistance to venetoclax appeared related (at least in part) to upregulation of anti-apoptotic members Mcl-1 and Bcl-xL, acquired Bcl-2 mutations, and the post-translational phosphorylation of Bcl-2 [61]. Thus, rational combinations of venetoclax with other agents were expected to produce a better clinical response [61]. In 2018, FDA expanded venetoclax’s market authorization so that it could be combined with the anti-CD20 rituximab for the treatment of patients whose CLL had progressed after a single course of treatment, regardless of their 17p deletion status [61]. When combined with rituximab or obinituzumab (also an anti-CD20 agent), venetoclax is now an approved standard of care for treatment- and relapsed CLL disease (Figure 2a and Table 2) [30]. Further, many clinical trials have combined venetoclax with other chemotherapeutic agents in CLL; for recent reviews, see [60,63]. Three ongoing phase II trials are evaluating the venetoclax + ibrutinib combination in treatment-naive CLL patients (NCT02910583) [64] and patients with relapsed/refractory (R/R) CLL/small lymphocytic lymphoma (SLL) (NCT0275689) [65], and the venetoclax + ibrutinib + obinutuzumab combination in treatment-naive CLL patients with a p53 deletion (17p-) and/or mutation (NCT02758665).
Bcl-2 is expressed strongly and constitutively in most B-NHL (Table 1). In this context, venetoclax has also been evaluated in patients with MM, FL, MCL, WM, MZL or even B-ALL or DLBCL (where ≈30% of tumors express high levels of Bcl-2) [3,60]. While venetoclax exhibited marked single-agent activity in MCL and WM, its outcome in treating the other B neoplasms was not as satisfactory [3,60,66]. Many clinical trials have started to evaluate various combination regimens of venetoclax in NHL and B-ALL (reviewed in [3,30,59,60,61,63]). Two phase II/III trials are evaluating the efficacy of venetoclax + ibrutinib in patients with relapsed MCL (NCT03112174) [67] and in WM with a specific MYD88 gene mutation (NCT04273139). In 2018, a phase I clinical trial of venetoclax + lenalidomide + rituximab started in patients with previously untreated MCL (NCT03523975). Unexpectedly, a phase III trial of venetoclax in R/R MM patients receiving bortezomib and dexamethasone as their standard therapy (NCT02755597) was suspended in 2022 [68]. A phase II study (NCT02055820) showed that venetoclax was efficacious and safe when combined with R-CHOP in DLBCL patients expressing high levels of Bcl-2 protein [69]. There are few trials of combinations of venetoclax with other targeted agents in FL, MZL and B-ALL [60]. With regard to B-ALL, the Bcl-2 and Bcl-xL dependence of cell lines and primary cells has prompted clinical trials of BCL2 antagonists as single agents or combination therapies [70,71]. A retrospective study confirmed the safety and efficacy of venetoclax when compared with conventional treatments of ALL in pediatric patients [72]. In a phase I study (NCT03181126), a combination of venetoclax with low-dose navitoclax and chemotherapy was well tolerated in both pediatric and adult patients with R/R B/T-ALL [73]. Two phase I/II trials (NCT03236857, NCT04029688) of venetoclax alone or combined with chemotherapeutics are being initiated for pediatric patients with relapsed B/T-ALL [74,75]. As a whole, these clinical trial data suggest that the addition of venetoclax to conventional chemotherapies improves response rates for NHL and B-ALL.
Treatment with venetoclax can be impeded by frequent gastrointestinal adverse events, neutropenia, and the emergence of specific, resistance-inducing BCL2 mutations [30,60]. Recently, two new Bcl-2 inhibitors were designed and developed: BGB-11417 targets Bcl-2 only, whereas lisaftoclax/APG-2575 targets Bcl-2 and Bcl-xL. In phase I trials, both compounds were found to be safe and effective in CLL/SLL and B-NHL (Table 3) [76,77]. These findings warrant further clinical investigation.
The anti-apoptotic protein Mcl-1 is a major factor in resistance to cancer chemotherapies and contributes to the onset of venetoclax resistance in some Mcl-1-overexpressing tumor cells [78]. Since 2012, many Mcl-1 inhibitors have been developed [79,80,81,82,83,84]. AMG176 [81], AMG-397 [82], and AZD5991 [84] were suspended due to the occurrence of serious adverse events in phase I trials for hematological malignancies, while the trial of ABBV-467 in R/R MM was terminated in 2021 for strategic reasons. A phase I study is now evaluating a novel Mcl-1 inhibitor (PRT1419) in patients with R/R MM and NHL (Table 3). Interestingly, the combination of another Mcl-1 inhibitor (S64315, also referred to as MIK665) with azacytidine is starting to be assessed in patients with acute myeloid leukemia (AML) [83]; the clinical value of S64315 in B lymphoid malignancies has yet to be assessed.
Table 2. Current treatments of B cell malignancies with FDA-approved drugs targeting Bcl-2, BTK, PI3K and TAAs (CD19/CD20/CD22/CD38), alone or in combination and as first- or second-line therapies.
Table 2. Current treatments of B cell malignancies with FDA-approved drugs targeting Bcl-2, BTK, PI3K and TAAs (CD19/CD20/CD22/CD38), alone or in combination and as first- or second-line therapies.
B Cell MalignancyCurrent Treatment OptionsReferences
B-ALLConventional chemotherapies *
Tyrosine kinase inhibitors imatinib or dasatinib for patients with Philadelphia chromosome-positive
Blinatumomab
Tisagenlecleucel
Inotuzumab ozogamycin (adults)
[85,86,87,88,89]
CLL/SLLFludarabine, cyclophosphamide and rituximab (FCR)
Bendamustine and rituximab (BR)
Rituximab and chlorambucil (RCb)
Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP)
Obinutuzumab and chlorambucil
Ibrutinib or acalabrutinib, alone
Idelalisib alone and with rituximab
Venetoclax + rituximab (or obinituzumab)
[60,90,91,92,93,94,95,96,97]
HCLPurine analogs (cladribine, pentostatin) alone and with rituximab
Moxetumomab pasudotox
[98,99]
MMConventional chemotherapies **
Daratumumab (or isatuximab) + lenalidomide (or pomalidomide) + dexamethasone
Belantamab mafodotin
[34,49,100,101,102]
FLBR
Rituximab, cyclophosphamide, vincristine, and prednisone (R-CVP)
R-CHOP
Rituximab + hyaluronidase human
Rituximab + lenalidomide
Obinutuzumab + bendamustine
Idelalisib or copanlisib alone
Ibritumomab tiuxetan (Yttrium-90)
Axicabtagene ciloleucel
[103,104,105,106,107]
MCLR-CVP, R-CHOP
Rituximab + lenalidomide
Rituximab + bortezomib
Ibrutinib or acalabrutinib or zanubrutinib, alone
Ibrutinib + rituximab
[53,108,109]
MZLR-CVP, R-CHOP, FCR, BR
Fludarabine and rituximab (FR)
Rituximab + lenalidomide
Ibrutinib or zanubrutinib
[54,110,111,112]
DLBCLR-CHOP
R-CHOP + etoposide
Rituximab
Tafasitamab + lenalidomide
Loncastuximab teserine
Tisagenleucel
Axicabtagene ciloleucel
[89,113,114,115,116,117]
WMAlkylating drugs and proteasome inhibitors both in combination with rituximab
Ibrutinib or acalabrutinib or zanubrutinib, alone and with rituximab
[59,118,119,120,121,122]
* Various combinations of vincristine, dexamethasone or prednisone, and an anthracycline drug (doxorubicin or daunorubicin); fludarabine, cytarabine and G-CSF; high-dose cytarabine; cytarabine and mitoxantrone; bortezomib. ** Proteasome inhibitors (bortezomib, carfilzomib, and ixazomib), immunomodulatory drugs (lenalidomide, pomalidomide, and thalidomide), alkylating agents (cyclophosphamide and bendamustine), steroids (prednisone and dexamethasone), a nuclear export inhibitor (selinexor), and bisphosphonates, alone or in various combinations. R-CHOP, rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone. The FDA-approved drugs listed in Figure 2a are written in bold type.
Table 3. Selected active or recruiting clinical trials of novel inhibitors of BCL2/BTK/PI3K proteins or TAAs (CD19/CD20/CD22/CD38), evaluated alone or in combination as first- or second-line therapies for B cell malignancies.
Table 3. Selected active or recruiting clinical trials of novel inhibitors of BCL2/BTK/PI3K proteins or TAAs (CD19/CD20/CD22/CD38), evaluated alone or in combination as first- or second-line therapies for B cell malignancies.
DrugClassDisease SettingPhaseStudyReferences
BGB-11417Bcl-2 inhibitorCLL/SLL,
B-NHL
INCT04277637
Recruiting, 2020–2023
Alone or combined with
zanubrutinib
[76]
Lisaftoclax/
APG-2575
Bcl-2/Bcl-xL inhibitorCLL/SLL, MZL, MCL


CLL/SLL

I



Ib/II

NCT03913949
Recruiting, 2019–2023

NCT04494503
Recruiting, 2020–2023
Alone or combined with
rituximab or ibrutinib
[77]



[123]
PRT1419Mcl-1 inhibitorR/R MM
R/R B-NHL
INCT04543305
Active, 2020–2022
[124]
S64351/
MIK-665
Mcl-1 inhibitorAMLI/IINCT04629443
Recruiting, 2020–2024
+ azacytidine
Orelabrutinib
/ICP-022
BTK inhibitorMCL
CLL/SLL


FL, MZL, MCL, CLL/SLL


R/R MZL, R/R WM,
DLBCL
I/II



I/II



II
NCT03494179
NCT03493217
Active, 2018–2022

NCT04014205
Recruiting, 2019-

NCT03797456
NCT04440059
NCT04438005
Recruiting, 2020-
[125]









Nemtabrutinib/
MK-1026/ARQ-531
BTK inhibitor CLL/SLL, MCL, R/R MZL R/R FL, R/R WMIINCT04728893
Recruiting, 2021–2027
Pirtobrutinib/
LOX-305
BK inhibitorPreviously treated CLL/SLL, WM, MCL, MZL


Untreated CLL/SLL



Previously treated CLL/SLL
I/II




III



III

NCT03740529
Recruiting, 2018–2023
Combined with venetoclax + rituximab


NCT05023980
Recruiting 2021-

NCT04965493
Recruiting, 2021-
Combined with venetoclax + rituximab
Parsaclisib/INCB500465/
IBI-376
PI3Kδ inhibitorCLL, DLBCLI/IINCT04809467
Recruiting, 2021–2023
Alone or combined with
tafasitamab
Zandelisib/PWT143/ME-401PI3Kδ inhibitorR/R FL,
R/R MZL



R/R CLL
III




II
NCT04745832
Recruiting, 2021–2031
Combined with rituximab

NCT05209308
Recruiting, 2022–2026
Combined with rituximab + venetoclax
[126]





BGB-10188PI3Kδ inhibitorR/R CLL, FL, MCL, DLBCL, MZLI/IINCT04282018
Recruiting, 2020–2025
Alone or combined with zanubrutinib and tiselizumab
Umbralisib/
TGR-1202
Dual PI3Kδ/CK1ε
inhibitor
R/R CLL, R/R MCL



R/R CLL, R/R B-NHL

I/Ib



I

NCT02268851
Active, 2014–2023
Combined with ibrutinib

NCT03283137
Active, 2017–2024
Combined with pembrolizumab
[127]





Mosunetuzumab/
BTC4465A
BiTE anti-CD20/CD3 CLL, B-NHL




B-NHL



I/II




I/II



NCT02500407
Recruiting, 2015-
Alone or combined with atezolizumab (anti-PD-L1)

NCT03671018
Recruiting, 2018-
Alone or combined with
polatuzumab
(anti-CD79B-monomethyl
auristatin E)
[128]











Odronextamab/
REGN1979
BiTE anti-CD20/CD3R/R DLBCL, FL, MZL, WMI/IINCT02651662
Active, 2016–2026
Alone or
combined with cemiplimab (anti-PD-1)
NCT03888105
Recruiting, 2019–2028
[129]





Epcoritamab/
GEN301
BiTE duoBody-anti-CD20/CD3R/R or progressive
DLBCL, MCL, FL, MZL, SLL

R/R DLBCL


R/R CLL
I/II


III


I/II
NCT03625037
Recruiting, 2018–2024

NCT04628494
Recruiting, 2020–2024

NCT04623541
Recruiting, 2020–2024
[130,131]





Glofitamab (RO7082859)BiTE anti-CD20/CD3 R/R B-NHLI/II





Ib/II
NCT03075696
Recruiting, 2017–2023
Alone or combined with
obinutzumab

NCT03533283
Recruiting, 2018–2024
Alone or combined with atezolizumab, obinutuzumab, tocilizumab polatuzumab vedotin
[132]







Tafasitamab/
MOR208/
Anti-CD19R/R FL, R/R MZL





DLBCL
III





III
NCT04680052
Recruiting, 2020-
Combined with rituximab + lenalidomide compared with rituximab + lenalidomide

NCT04824092
Recruiting, 2021–2026
Combined with lenalidomide + R-CHOP





[133]
CAR-20.19-TAnti-CD19/CD20 CAR-T/CD28/
4-1BB/CD3ζ
CLL/SLL, R/R B-NHLINCT03019055
Active, 2017–2022
[134]
CD19-CAR-NK
using blood cord NKs
Anti-CD19 CAR-NK/OX40/CD3ζ/IL-15B-ALL CLL/SLL,
R/R B-NHL
I/IINCT03056339
Active, 2017–2022
after treatment with fludarabine/cyclophosphamide lymphodepletion
[135,136]
NKX019
using peripheral blood NKs
Anti-CD19 CAR-NK/OX40/
CD3ζ/IL-15
B-ALL CLL/SLL,
R/R B-NHL
INCT05020678
Recruiting 2021–2023
[137]
C-CAR066/CBM.20-CAR-TAnti-CD20 CAR-T NHLs resistant to rituximab or anti-CD19 CAR-TINCT04036019
Recruiting, 2019–2021
NCT04316624
Recruiting, 2020-
[138]
Inotuzumab ozogamicinADC anti-CD22- calicheamicinB-ALL, B-NHLIIINCT03959085
Recruiting, 2019–2029
post chemotherapy
JNJ-75348780BiTE anti-CD22/CD3CLL, B-NHLINCT04540796
Recruiting, 2020–2024
CD22-CARAnti-CD22 CAR-T/CD8/4-1BB/CD3ζR/R B-ALL,
B-NHL
INCT02315612
Recruiting, 2014–2040
[139]
LV20.19Bi anti-CD19/CD22 CAR-T/4-1BB/CD3ζCLL/SLL, B-NHLINCT03019055
Active, 2017–2022
[134,140]
AUTO3Bi anti-CD19 (OX40 costim)/CD22 (4-1BB costim) CAR-TR/R DLBCLI/IINCT03287817
Active 2017-
[141]
CD19-22.BB.z-CARBi anti-CD19/
CD22 CAR-T
R/R B-ALL, DLBCL





Pretreated B-ALL
I





I
NCT03233854
Recruiting, 2017–2025
Combined with NKTR-255 (IL-15 receptor agonist)

NCT03448393
Recruiting, 2018–2040
following cyclophosphamide/fludarabine treatment
[142]




[143]
CD20-22 CAR Bi anti-CD20/CD22 CAR-TB cell malignanciesINCT04283006
Recruiting, 2020–2028
Felzartamab/
MOR202/TJ202
Anti-CD38R/R MMII




III
NCT03860038
Active, 2019–2022
Combined with
dexamethasone

NCT03952091
Active, 2019–2022
Combined with lenalidomide + dexamethasone
TAK-079Anti-CD38R/R MM




MM


I/IIa




I

NCT03499280
Completed, 2018–2022
Alone or combined with pomalidomide and dexamethasone

NCT03984097
Active, 2019–2023
Combined with lenalidomide + dexamethasone
TAK-573/
modakafusp
ADC anti-CD38 delivering attenuated IFN-α2b R/R MMI/IINCT03215030
Recruiting, 2017-
Alone or combined with dexamethasone
[144]
TAK-169/
MT-0169
ADC anti-CD38 delivering Shiga-like toxin R/R B-NHLINCT04017130
Recruiting, 2019-
[145]
ISB 1342/
GBR 1342
BiTE anti-CD38/CD3R/R MMINCT03309111
Recruiting 2017–2024

2.2. BTK Inhibitors

Of the many BTK inhibitors developed in the last 10 years, three (ibrutinib/PCI-32765, acalabrutinib/ACP 196, and zanubrutinib/BGB 3111) are now used in the clinic to treat B cell malignancies (Figure 2a and Table 2). Ibrutinib and acalabrutinib are authorized for untreated and R/R CLL/SLL and CLL with 17p deletion [146,147,148,149], ibrutinib and zanubrutinib are authorized for R/R MZL patients who have received at least one line of an anti-CD20-based regimen [149], and ibrutinib, acalabrutinib and zanubrutinib are authorized for WM patients and for MCL patients who have received at least one line of treatment [119,147,148,149,150]. These three agents are orally administered alone or in combination with an anti-CD20 mAb such as rituximab (Table 2). Acalabrutinib and zanubrutinib are better tolerated than ibrutinib, although the first two drugs have not been on the market as long as the latter [149]. All three drugs inhibited BTK irreversibly by forming a covalent bond with cysteine-481 (Cys-481) in the active site [28,151]. The first-generation BTK inhibitor ibrutinib has off-target kinase inhibition activities [28], whereas the second-generation BTK inhibitors acalabrutinib and zanubrutinib are more selective for BTK [28,149].
Several completed phase I trials have demonstrated the safety of ibrutinib and acalabrutinib in R/R MM, either alone or combined with other therapeutics [152]. A phase III trial (NCT01855750) showed a survival benefit of the addition of ibrutinib to R-CHOP chemotherapy in younger patients with DLBCL [153], while a phase II study (NCT01779791) did not find any benefit of ibrutinib in R/R FL [154]. In a phase III trial (NCT03734016), zanubrutinib significantly improved response rates and delayed disease progression in patients with R/R CLL/SLL (relative to ibrutinib) and did so with less toxicity [155]. The efficacy and safety of zanubrutinib are being evaluated in treatment-naive CLL/SLL patients with and without a 17p deletion (NCT03336333). Adding zanubrutinib to obinutuzumab improves responses and progression-free survival in R/R FL (NCT03332017, 2017–2023) [156]. In a phase II study (NCT03145064), zanubrutinib monotherapy led to modest responses in R/R DLBCL patients with both CD79B and MYD88 mutations [157].
The resistance of lymphoid tumors to treatment with first- and second-generation BTK inhibitors and subsequent relapse appear to be in part related to the acquisition of mutations in BTK [149,158]. The most common is the Cys-481 to serine (C481S) mutation, which disrupts inhibitor-BTK binding [149]. To overcome this resistance problem, third-generation noncovalent BTK inhibitors have been developed to inhibit the kinase activities of both BTK and BTKC481S [28,149]. These drugs include vecabrutinib/SNS-062, fenebrutinib/GDC-0853, orelabrutinib/ICP-022, nemtabrutinib/MK1026/ARQ-531, and pirtobrutinib/LOX-305. The completed phase I trials with vecabrutinib in CLL/SLL and B-NHL (NCT03037645) and with februnitib in R/R NHL and CLL (NCT01991184) showed that these drugs were well tolerated but exhibited low levels of antitumor activity; hence, the studies were discontinued [159]. Orelabrutinib, pirtobrutinib, and nemtabrutinib are undergoing clinical development as monotherapies or combination therapies in most B cell malignancies (Table 3).

2.3. PI3K Inhibitors

PI3K isoforms (PI3Kα, PI3Kβ, PI3Kδ and PI3Kγ) are commonly found in B cell malignancies and are activated by BCR signaling [160,161]. This is why targeting the PI3K pathway has promise in treating B cell malignancies. At present, two PI3K inhibitors are used in routine clinical practice for the treatment of patients with CLL and FL: idelalisib (an orally administered PI3Kδ inhibitor) and copanlisib (an intravenously administered pan-PI3K inhibitor) [162,163] (Figure 2a and Table 2). As the first-in-class PI3Kδ inhibitor, idelalisib showed efficacy in patients with a 17p deletion and p53 mutation, unmutated IGHV status, and R/R CLL [161]. The drug is currently used in combination with rituximab (Table 2) [164]. Idelalisib and copanlisib have been approved for the treatment of R/R FL in patients having received two or three lines of treatment (Table 2) [107,164,165,166]. A recent, retrospective, Italian multicenter study confirmed the effectiveness of idelalisib in R/R FL patients treated in routine clinical practice [167]. Compared with idelalisib, copanlisib was associated with a higher complete remission rate and a more favorable safety profile [107]. It should be noted that the oral dual PI3Kδ/γ inhibitor duvelisib (initially approved in 2018 for the treatment of patients with R/R FL having received at least lines of systemic treatment [168]) was withdrawn from the market by its developer Secura Bio in December 2021, following the occurrence of fatal and/or serious infections [169].
Despite the recent withdrawal of duvelisib in the indication of FL, several clinical trials in CLL/SLL are still evaluating duvelisib as combination treatments and with different dosing regimens [161,162]. A phase III trial (NCT02004522) of duvelisib vs. ofatumumab (an anti-CD20) in patients with R/R CLL/SLL was terminated in 2021 after the occurrence of serious adverse events, including severe infections and diarrhea/colitis [161]. Clinical evaluations of the efficacy of idelalisib as a monotherapy and of duvelisib combined with anti-CD20 (obinutuzumab) in R/R MM were recently reviewed by Grimont et al. [119]. Idelalisib is associated with significant hepatotoxicity, which may limit the drug’s use in WM [118]. Four phase I/II studies are evaluating copanlisib as part of a combination therapy for CLL, FL, MZL and DLBCL (NCT03884998), DLBCL (NCT03484819), untreated FL (NCT03789240), and DLBCL and R/R grade 3b FL (NCT03789240) (reviewed in [107,164]). In an ongoing phase III trial (NCT02367040), copanlisib was found to be safe when combined with rituximab for the treatment of R/R NHL [170].
Given the potentially limiting toxicities reported for idelalisib and copanlisib [161], further improvements are required to develop new, safer, and more selective PI3K inhibitors. Many PI3K inhibitors have been developed over the last five years, and some of them are being tested in the clinic as monotherapies or combination therapies (reviewed in [12,160,161]). For example, the oral PI3Kδ inhibitor parsaclisib (used alone or in combination with chemotherapy) demonstrated antitumor activity and an acceptable safety profile in R/R B-NHL (the phase II NCT02998476 and phase III NCT02018861 both completed in 2021) [171,172]. Following a business decision in February 2022, Incyte withdrew parsaclisib as a treatment for patients with R/R MCL, MZL and FL [173]. However, a combination of parsaclisib with the anti-CD19 mAb tafasitamab is still being evaluated in patients with R/R-CLL and DLBCL (Table 3). Furthermore, two potent, selective PI3Kδ inhibitors (zandelisib and BGB-10188) are in clinical development for the treatment of patients with R/R CLL and B-NHL. Zandelisib (alone or in combination with rituximab) was found to be well tolerated in all R/R neoplasms CLL/SLL, R/R FL, MZL and DLBCL (NCT02914938) [174]. A multicenter, randomized phase III study is now investigating the safety and efficacy of the zandelisib + rituximab combination vs. standard immunochemotherapy in patients with R/R FL or MZL (Table 3) [126], while a phase II trial has been initiated with a zandelisib + rituximab + venetoclax combination in patients with R/R CLL (Table 3). In a phase I/II study, BGB-10188 is being evaluated as a monotherapy and a combination therapy with zanubrutinib and tislelizumab (an anti-PD1) in patients with CLL and B-NHL (MCL, FL, DLBCL, and MZL) (Table 3). Lastly, umbralisib/TGR1202 inhibits both PI3Kδ and casein kinase 1ε (CK1ε); the latter is involved in the translation of the c-Myc oncogene and the regulation of the Wnt pathway [175]. Umbralisib is associated with less immune-mediated toxicity than the other PI3Kδ inhibitors (because it disables WNT signaling by inhibiting CK1ε) and exerts less detrimental effects on regulatory T cells’ immunosuppressive functions [175]. Umbralisib was found to be efficacious and safe as a monotherapy for patients with R/R CLL and B-NHL [176] and for CLL patients who do not tolerate BTK or PI3Kδ inhibitor therapy [177]. The drug’s favorable toxicity profile was confirmed when it was combined with ibrutinib in patients with R/R CLL and MCL [127] or with ublituximab (an anti-CD20) in patients with CLL/SLL and B-NHL [178]. In January 2022, however, the FDA placed a partial clinical hold on studies of umbralisib and ublituximab [160,161] in an indication of CLL or B-NHL [179]. Studies of a combination of umbralisib with ibrutinib or pembrolizumab (an anti-PD1) in patients with R/R CLL and R/R B-NHL are ongoing (Table 3).

2.4. mAbs and CAR-T Cells

Antibody-based cancer treatments have developed quickly over the past 25 years. Following on from the first generation of therapeutic murine mAbs (-momabs), a variety of second-generation mAbs were engineered: chimeric mAbs (-ximabs), humanized mAbs (-zumabs), and human mAbs (-umabs) [180]. Chimeric mAbs possess murine variable regions, whereas the rest of the Ab is of human origin. Humanized mAbs retain murine hypervariable segments, whereas fully human mAbs have a native Fc region [180]. Third-generation mAbs have an engineered Fc region, which is designed to improve therapeutic performance by adapting their effector functions [180]. The main effector pathways commonly employed by mAbs are complement-dependent cytotoxicity (CDC), Ab-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and programmed cell death (PCD) [181,182].
To enhance their therapeutic value, mAbs have also been conjugated with toxins (giving ADCs), radioisotopes (giving ARCs), immunomodulatory cytokines, and costimulatory molecules (giving CAR-T cells) [180,182,183,184,185]. An ADC combines selectivity and cytotoxic potency and so can target a given TAA with high specificity [183,186]. After binding to a TAA, ADCs enter the cell via receptor-mediated endocytosis and release the lethal drug. ADCs can also activate ADCC and ADCP signaling by immune effector cells [186]. Multivalent Abs simultaneously target a TAA and an activating receptor on the effector cell (typically a T cell) [187]. Most engineered, multispecific Abs lack an Fc region and therefore do not mediate CDC or ADCC [184]. Bispecific T-cell engagers (BiTEs) are bispecific, single-chain Abs that specifically recognize the T cell antigen CD3 and a TAA, which also stimulate cytotoxic T cells for targeted tumor cell lysis [184,185]. CAR-T therapy is based on antibody recognition of TAAs [188,189]. CAR-T cells are cytotoxic T cells that have been modified genetically to express an artificial chimeric antigen receptor (CAR). The CAR comprises an extracellular target-binding domain (composed of a single-chain variable fragment (scFv) from antibody light- and heavy-chain variable regions and which bind to the TAA), a spacer/hinge region, transmembrane domains, and intracellular domains (comprising two or more costimulatory domains (CD28 and/or 4-1BB/CD137/TNF-RSF9) and a T cell signaling domain (with CD3ξ) [14,190,191]. The first generation of CAR constructs included CD3ξ; the second generation included CD28 and CD3ξ, or 4-1BB and CD3ξ; the third generation included CD28, 4-1BB, and CD3ξ; the fourth generation included a cytokine transgene initiated through NFAT signaling, and the fifth generation included IL-2Rβ (to initiate JAK/STAT signaling) [14,190,191]. Following expansion in the laboratory and then infusion into the patient, CAR-T cells bind to their cognate targets on tumor cells through their extracellular domain and initiate signal transduction that triggers the effector functions of host cytotoxic T cells, which then kill the tumor cells [188,189].

2.4.1. CD20 mAbs and CD20-Targeting CAR-T

Within the last 20 years, a variety of strategies have been used to develop a large number of anti-CD20 mAbs. The FDA’s approval of rituximab in 1997 marked a breakthrough in the treatment of B cell malignancies (Figure 2a). Rituximab is still part of the standard of care for most B cell tumors (Table 2). Rituximab engages the main effector pathways employed by mAbs (i.e., CDC, ADCC, and ADCP) and even directly activates PCD [182,192]. Many phase I/II/III trials have evaluated the combination of other novel drugs (including ibrutinib, idelalisib, acalabrutinib, venetoclax, ipilimumab (an anti-CTLA4), chromatin modulators, and other drugs) with rituximab in B cell malignancies [15,193]. Based on the efficacy, relative safety and ubiquitous use of rituximab in B cell malignancies, attempts have been made to develop more efficacious and/or less toxic CD20 mAbs [15,184,185,194,195]. Indeed, the CD20 mAbs obinutuzumab and ibritumomab tiuxetan (radiolabeled with 90Yttrium) are now used to treat CLL and FL (Figure 2a and Table 2), and four BiTE anti-CD20/CD3 mAbs (mosunetuzumab, odronextamab, glofitamab and epcoritamab) are in clinical development (Figure 2b and Table 3) [128,130,131].
Obinutuzumab’s mechanisms of action are similar to those of rituximab [182,195]. 90Y-ibritumomab binds to the CD20 antigen on tumor B cells, and the long range of β particles from the 90Y means that neighboring tumor cells can be killed without direct antibody binding [195]. Compared with rituximab in the induction therapy of B-NHL, obinutuzumab was associated with significantly greater progression-free survival but a greater incidence of adverse events, and 90Y-ibritumomab was associated with a greater overall response rate [33].
Mosunetuzumab, odronextamab and glofitamab monotherapies have already show manageable safety profiles and given lasting responses in heavily pretreated patients with R/R B-NHL (Table 3) [128,132,196]. Compared with intravenously infused mosunetuzumab, odronextamab or glofitamab, the subcutaneously administered epcoritamab is usefully associated with (i) lower peak cytokine levels, (ii) a lower treatment burden for patients, and (iii) less resource use at the treatment facility [130,131]. Mosunetuzumab is currently in phase I/II trials, where it is combined with mAbs targeting the programmed cell death protein 1 (PD-1) immune checkpoint, the latter’s ligand (programmed death-ligand 1, PD-L1), or CD79B (an antigen associated with the BCR; see Figure 1b) (Table 3). Other novel BiTE anti-CD20/CD3 mAbs are being evaluated in phase I trials [184].
Several clinical studies have focused on the potential of autologous, first-generation anti-CD20 CAR-T cell therapies in patients with B cell malignancies (reviewed in [138,197,198]). Two phase I clinical trials are underway to evaluate the efficacy of C-CAR066 (a novel, second-generation, anti-CD20 CAR-T cell therapy) in patients with R/R NHL (including cases of DLBCL) after the failure of rituximab or CD19 CAR-T therapy (Table 3) [138]. All the enrolled B-NHL patients who were previously R/R to rituximab achieved various clinical responses, and the level of toxicity was tolerable [138].

2.4.2. CD19 mAbs and CD19-Targeting CAR-T

Many models based on CD19 mAbs have been built and evaluated in the treatment of B cell malignancies [89,199,200]. Several strategies have been implemented, and four types of CD19 mAbs are currently used to treat B-ALL, DLBCL and FL (Table 2): blinatumomab (a BiTE), tafasitamab (an Fc–engineered and Fab affinity-matured Ab), loncastuximab tesirine (ADC), and CAR-T therapies (lisocabtagene maraleucel, axicabtagene ciloleucel and tisagenlecleucel) (Table 2) [89,201,202,203,204,205,206].
By binding both CD19 on B cells and CD3 on T cells, blinatumomab triggers ADC-dependent cytotoxicity [89]. As the first FDA-approved BiTE for the treatment of B-ALL, blinatumomab (developed by Amgen) induces remissions in cases of R/R B-ALL that had failed to respond to other therapies (Table 2) [201,207]. However, blinatumomab has a short half-life and must be continuously infused intravenously [201]. Of the many other CD19 mAbs developed [199], tafasitamab (MOR208) is combined with lenalidomide in the treatment of R/R DLBCL (Table 2) [115,116]. Tafasitamab mediates both tumor B cell apoptosis, ADCC and CDC [208,209]. The main adverse event associated with tafasitamab is bone marrow suppression, with anemia, neutropenia, and thrombocytopenia [115]. At present, two phase III clinical trials are recruiting patients; a combination of tafasitamab and first line-chemotherapeutic agents will be used to treat DLBCL (NCT04824092) [14,89,133] or R/R FL and R/R MZL (NCT04680052) [14,89,133]. Loncastuximab tesirine (ADCT-402) is a humanized CD19 mAb linked to teserine (SG32499); lysosomal degradation delivers a pyrrobenzodiazepine dimer that crosslinks the tumor cell’s DNA [117,202]. Based on the phase II LOTIS-2 trial, loncastuximab tesirine was recently granted fast-track approval for the therapy of adult patients with R/R DLBCL and who have already received two or more lines of treatment (Table 2) [210]. An ongoing phase I clinical trial (NCT03684694) is investigating a combination of loncastuximab and ibrutinib in the treatment of patients with DLBCL and MCL.
The three CD19-CAR-T cell therapies on the market (tisagenlecleucel, axicabtagene ciloleucel, and lisocabtagene maraleucel) use autologous T cells [89,203,205,211]. CAR-T engagement of CD19+ tumor cells results in T-cell activation and proliferation, the secretion of inflammatory cytokines and chemokines, and thus tumor cell lysis [212]. Tisagenlecleucel is composed of an anti-CD19 scFv, a CD8-α hinge region, 4-1BB, and CD3ζ [213]. Axicabtagene ciloleucel contains an anti-CD19 scFv, an IgG4 hinge region, CD28, and CD3ζ [212]. Lisocabtagene maraleucel contains an anti-CD19 scFv, an IgG4 hinge region, and CD28, 4-1BB and CD3ζ domains [214]. Tisagenlecleucel has been approved for the treatment of children and young adults with R/R B-ALL (Table 2) [88,213,215,216,217]. In recent phase II/III studies of pediatric/young adult patients with Down-syndrome-associated ALL (NCT02435849, NCT02228096, and NCT03123939), tisagenlecleucel treatment was associated with high remission rates, manageable side-effects, and promising long-term outcomes [216]. From the third line of treatment onwards, both tisagenlecleucel and axicabtagene ciloleucel are currently recommended for patients with DLBCL who have failed autologous transplantation or have relapsed after two previous lines of treatment (Table 2) [32,212,215]. A recent retrospective evaluation of the safety and efficacy of axicabtagene ciloleucel and tisagenlecleucel in DLBCL outside the setting of a clinical trial found that although the two cohorts did not differ significantly with regard to any of the baseline characteristics, cytokine release syndrome and neurologic events were more frequent in the axicabtagene group [218]. An ongoing phase II trial is looking at tisagenleucel CAR-T cell therapy for FL (NCT04094311, 2019–2023). At present, there are three active phase II clinical trials evaluating axicabtagene alone in FL (NCT03105336) [219] and in combination with first line-chemotherapeutic agents (cyclophosphamide/fludarabine) in DLBCL (NCT03761056, NCT03391466) [220]. In 2022, lisocabtagene was approved as a second-line treatment of patients with R/R DLBCL and FL and who were not eligible (due to comorbidities or age) for hematopoietic stem cell transplantation (Table 2) [214]. Two phase I/II studies will evaluate the efficacy and safety of lisocabtagene in combination with other chemotherapeutic agents in R/R CLL/SLL (NCT03331198, with ibrutinib or venetoclax) [221], and high-risk DLBCL and FL (NCT03310619: lisocabtagene alone or combined with ibrutinib or durvalumab). Relative to standard care, the lisocabtagene + ibrutinib combination yielded significantly higher event-free survival and response rates in patients with R/R CLL/SLL [221]. Thus, CD19-CAR-T therapy appears to have promising outcomes and a tolerable safety profile in R/R B cell malignancies [222,223]. Further modifications of the CAR’s structure (resulting in second/third-generation anti-CD19/CAR-T agents) and the therapeutic strategy are being tested as treatments for B cell malignancies (reviewed in [31,223,224]).
Given that the emergence of CD19- tumor cells substantially increases the risk of relapse [32], another approach seeks to optimize single-chain, bispecific CARs (biCARs) that trigger robust cytotoxicity against tumor cells expressing either CD19 or CD20 [134,225,226]. Dual targeting may improve response rates and sustained responses while limiting antigen escape [134,225,226]. In the first-in-human biCAR phase I/IIa trial (NCT03097770, completed in 2020), a bispecific anti-CD19/CD20 tandem receptor (tanCAR7 with 4-1BB/CD3ζ) elicited a potent, lasting antitumor response in the absence of grade 3 or higher cell-related encephalopathy syndrome in patients with R/R NHL [226]. Similarly, another anti-CD19/anti-CD20 CAR-T (CAR-20.19-T) showed low toxicity and high efficacy as single agent in a phase I study with R/R B-NHL (Table 3) [134].
CAR-engineered natural killer (CAR-NK) cells constitute another CAR-based approach to cancer therapy [227,228]. The first anti-CD19 CAR-NK cell construct was composed of an anti-CD19 scFv, CD28, a costimulatory OX40 domain, CD3ζ, and a membrane-bound form of IL-15 [135]. The addition of IL-15 enhanced the proliferation, persistence and activity of NK cells in preclinical models [135]. In a phase I/II trial, HLA-mismatched anti-CD19 CAR-NK cells derived from cord blood were administered to patients with R/R B-NHL, CLL/SLL and B-ALL after standard fludarabine/cyclophosphamide lymphodepletion (Table 3) [135,136]. The majority of the patients responded to the CAR-NK treatment and did not develop serious adverse events [135,136]. More recently, a similar anti CD19-NK cell therapy (NKX019, using HLA-mismatched anti-CD19 CAR-NK cells obtained from peripheral blood) entered a phase I study as a monotherapy for patients with R/R B-NHL, CLL and B-ALL (Table 3) [137]. NKX019 treatment has already shown long-term NK cell persistence and potent antitumor activity [137]. Additional clinical trials for CAR-NK cells for the treatment of B cell malignancies are being set up [14,228].

2.4.3. CD22 mAbs and CD22-Targeting CAR-T

CD22-targeting mAbs have emerged as promising treatment options with proven therapeutic value [16,21,98]. Two ADC-triggering anti-CD22 mAbs (inotuzumab ozogamicin (CMC544) and moxetumomab pasudotox) have been approved for the treatment of adult patients with B-ALL and R/R HCL, respectively (Table 2) [99,229,230,231,232]. Inotuzumab ozogamicin combines an anti-CD22 mAb with the DNA-binding drug calicheamicin, which leads to DNA breakage and apoptosis [16,21]. Moxetumomab pasudotox is an anti-CD22 scFv conjugated to a truncated 38 kDa fragment of Pseudomonas exotoxin A (PE38) [233]. In a phase I/II study (NCT01371630, 2011–2025), a combination of inotuzumab ozogamicin with mini-hyperfractionated cyclophosphamide, vincristine, dexamethasone and (in some cases) blinatumomab was found to be highly effective in patients with B-ALL after their first relapse [234]. A phase II study (ITCC-059; EUDRACT 2016-000227-71; NTR5736) provided further evidence of inotuzumab’s activity in pediatric patients with R/R B-ALL [235]; the study is ongoing and is testing a combination of inotuzumab with chemotherapy in pediatric patients with R/R ALL. In an ongoing phase III randomized trial, inotuzumab ozogamicin is being tested as a first-line treatment with high-risk B-ALL and B-lymphoblastic lymphoma (Table 3). However, phase I/II studies of inotuzumab ozogamicin alone or in combination with rituximab for the treatment of B-NHL did not give promising results [236,237,238]. Based on a phase I study of patients with R/R ALL, moxetumomab pasudotox’s clinical activity appears to be moderate [231,239]. Only one phase I study with a BiTE construct targeting CD22 and CD3 (JNJ-75348780) is currently assessing patients with R/R NHL and R/R CLL (Table 3).
Over the last 10 years, many CD22-targeting CARs have been developed and tested in phase I/II clinical trials [16,240]. Two anti-CD22 CARs containing anti-CD22 scFv, IgG4 (or CD8), 4-1BB and CD3ζ domains gave high response rates in patients with B-ALL who had previously failed chemotherapy and/or CD19-targeting CAR-T treatment (Table 3) [139]. Moreover, many bispecific anti-CD19/CD22 CAR-T cells have been developed to prevent relapse by minimizing antigen escape and have been assessed in phase I studies for the treatment of B-cell malignancies [16]. The first anti-CD19/CD22 CAR-T cell construct (4-1BB-CD3ζ, LV20.19 CAR-T) contained a CD19-binding domain derived from a clinically active CD19-CAR, together with a CD22 binding domain [241]. In a phase I trial, LV20.19 showed low toxicity and high efficacy in R/R NHL and CLL (Table 3) [134]. On the same lines, the combination of another dual CD19/22-targeting CAR-T cell therapy (AUTO 3) with pembrolizumab led to complete remission in patients with R/R DLBCL (Table 3) [141]. A new anti-CD19/CD22-CAR-T construct (CD19-22.BB.z, alone or combined with an IL-15 receptor agonist) is being assessed in patients with R/R DLBCL or B-ALL in a phase I trial (Table 3) [142]. According to the initial results, almost all the patients with ALL achieved complete remission [142]. The results of a phase I trial recently confirmed the safety and efficacy of CD19.22.BB.z in heavily pretreated children and young adults with B-ALL (Table 3) [242]. On the basis of these preliminary data, it appears that anti-CD19/CD22 CAR-T cells may improve clinical responses by mitigating target antigen downregulation as a relapse mechanism. Another novel, bispecific anti-CD20/CD22 CAR-T is now being tested for safety in B-cell malignancies (Table 3). A phase I study of monotherapy with anti-CD22-NK cells is planned in patients with R/R NHL and who previously received anti-CD19 treatment (NCT03692767: not yet recruiting).

2.4.4. CD38 mAbs and CD38-Targeting CAR-T

Although the development of therapeutic anti-CD38 mAbs started in the 1990s and has progressed slowly since then, two such mAbs (daratumumab and isatuximab) are now used as combination chemotherapies for patients with R/R MM (Figure 2a and Table 2) [243,244,245,246]. In MM, the two Abs induce ADCC, CDC and ADCP as part of their antitumor mechanism of action [247,248]. Furthermore, isatuximab directly induces both lysosome-dependent death and caspase 3/7-dependent apoptosis of MM cells–even those bearing a p53 mutation [249]. Unlike daratumumab, isatuximab binds to a CD38 epitope that encompasses the CD38 ectoenzyme’s catalytic site, but it is not known whether isatuximab exerts its therapeutic effect by inhibiting CD38’s NADase activity [17]. Usefully, daratumumab is available in formulations for intravenous administration and subcutaneous administration [34,195]. One advantage of isatuximab over daratumumab is the need for less frequent dosing [34,195]. The most common adverse reactions to these two anti-CD38 Abs are neutropenia, pneumonia, upper respiratory tract infection, and diarrhea [34,195].
Following the observation of daratumumab’s and isotuximab’s efficacy in R/R MM, several clinical trials have evaluated the addition of one mAb or the other to standard-of-care regimens in newly diagnosed MM patients [250,251,252]. Based on these studies, the combination of daratumumab with bortezomib/thalidomide/dexamethasone (VRd) was recently approved by the FDA as treatment for these transplant-eligible patients [253]. Since then, two phase III trials (NCT03710603, NCT03652064) have compared the daratumumab-VRd combination with VRd in non-transplant-eligible patients and as an induction treatment before autologous stem cell transplantation in transplant-eligible patients. Other phase III trials are assessing the benefit of combining isatuximab with (i) lenalidomide + bortezomib + dexamethasone or lenalidomide + carfilzomib induction treatment in patients with newly diagnosed MM (NCT03617731, NCT03319667, and NCT04483739), (ii) carfilzomib in patients with R/R MM (NCT03275285, 2017–2023), and (iii) lenalidomide in patients with high-risk smoldering MM (NCT04270409).
Data on the efficacy of daratumumab and isatuximab in other B cell malignancies are scarce. Daratumumab has been tested in phase I trials on patients with CLL and other B-NHL. In CLL, the prognostic value of CD38 expression in patients is well proven (Table 1), and preclinical studies (ex vivo experiments and CLL mouse models) have demonstrated the antitumor efficacy of daratumumab monotherapy or combination therapy [254,255]. The combination of daratumumab with ibrutinib in CLL patients with a poor prognosis (e.g., 17p deletion and/or p53 mutation) is now being studied (NCT03447808 and NCT03734198). The putative efficacy of daratumumab in B-ALL and other types of NHL has not yet been tested. A phase I/II study of the safety and tolerability of a combination of isatuximab with the anti-PD1 cemiplimab in patients with DLBCL started in 2018 (NCT03769181). The fact that CD38 is strongly expressed by some tumor cell subsets in pediatric patients with B-ALL [256] makes this antigen a potential target for the treatment of ALL in children. Isatuximab has significant antitumor activity in vitro and in ALL xenograft models, with robust ADCC- and ADCP-mediated effects [256]. A now-recruiting phase II multicenter study is evaluating the antitumor activity, safety, and pharmacokinetics of isatuximab combined with standard salvage chemotherapies in children after one or two relapses of B-ALL (NCT03860844) [257].
Along with daratumumab and isatuximab, several novel CD38-targeting strategies are being developed (mostly for MM); these include the use of naked anti-CD38 mAbs, anti-CD38 ADCs, anti-CD38/CD3 BiTEs, and CD38-targeting CAR-T cells [195,247,251,258]. Felzartamab (MOR202; TJ202) and TAK-079 are both fully human IgG1 mAbs [251,259]. Although felzartamab showed promising efficacy in MM (both alone and combined with immunomodulators; NCT01421186) [260], the drug’s sponsor (MorphoSys AG) unexpectedly stopped this development in 2020 [259]. However, two ongoing trials are evaluating the effect of a combination of felzartamab and conventional therapies in patients with R/R MM (Table 3). The results of a multicenter phase I study showed that monotherapy with subcutaneously administered TAK-079 is safe and generally well tolerated in patients with R/R MM (Table 3) [261]. Two other clinical trials are evaluating a combination of TAK-079 with polalidomide or lenalidomide + dexamethasone in R/R MM and newly diagnosed MM (Table 3).
Two ADC-triggering anti-CD38 drugs (TAK-573 and TAK-169) have recently entered clinical development in the treatment of R/R NHL (Table 3) [144,145]. TAK-573 (modakafsup) is a CD38 mAb fused to attenuated interferon (IFN)-α; it demonstrates potent anti-MM activity in patients with R/R MM [144]. TAK-169 is an engineered Ab with a deimmunized form of the ribosome-inactivating Shiga-like toxin A subunit genetically fused to a CD38-binding Ab fragment [145]. TAK-169 is internalized by CD38+ MM cells, inactivates ribosomes, abrogates protein synthesis and thus kills cells directly [145]. The full results of these clinical trials are eagerly awaited.
Among the many humanized anti-CD38/CD3 BiTEs having been through preclinical development, AMG-424 and ISB 1342 (GBR 1342) have shown strong antitumor effects in MM models [262,263]. A phase I study (NCT03445663) of AMG424 in R/R MM began in 2018 but was terminated by the sponsor for business reasons in 2020. Since ISB 1342 contains a full Fc domain with a reduced effector function, its mechanism of tumor cell killing critically relies on the engagement and activation of T lymphocytes [263]. ISB 1342 is currently in phase I trial for the treatment of patients with MM who have relapsed or who do not respond to standard therapies (e.g., proteasome inhibitors, immunomodulators, and daratumumab) (Table 3).
As is the case for CD19, CD20, and CD22, CD38 is an attractive target for CAR-T therapy. However, given the widespread expression of CD38 (e.g., in plasma cells, precursor B cells, T cells, NK cells, myeloid precursor cells, and various organs), anti-CD38 CAR-T cell constructs can induce various syndromic adverse events [251]. For instance, the administration of anti-CD38 CAR-T cells to a patient with relapsed B-ALL (after the failure of bispecific anti-CD19/CD22 CAR-T cell treatment) was associated with target-mediated toxicity and severe cytokine release syndrome [264]. Some novel anti-CD38 CAR-T cells have been developed with a modified light chain and a high-affinity scFv [251]. A phase I study has evaluated the safety and efficacy of a novel CD38 A2 CAR-T cells construct in patients with R/R MM (NCT03464916, completed in 2022) but it is not known whether this construct has been efficacious in phase II/III trials. The strategies assessed in MM could also be investigated in the treatment of other CD38+ lymphoid tumors, including poor-prognosis CLL/SLL, HCL, and other B-NHL. Lastly, the promising preclinical efficacy seen with CD38-targeting NK cells suggests that clinical development is warranted [265,266].

3. Clinical Trials of Novel Anti-TAA Inhibitors in the Treatment of B Cell Malignancies

3.1. B Cell Maturation Antigen (BCMA) and B-Cell Activation Factor Receptor (BAFF-R)

BCMA (also known as tumor necrosis factor receptor superfamily member 17/TNFRS17) is a transmembrane protein that contains cysteine-rich extracellular domains and lacks a signal peptide [267]. Along with the related TNFR superfamily member BAFF-R, BCMA critically regulates B cell proliferation, survival and differentiation into plasma cells [102,267,268]. BCMA is expressed at significantly higher levels by MM cells (Table 1) than by normal tissues (except for normal plasma cells) [269]. Thus, BCMA has become an important therapeutic target in MM, with four treatment modalities in development: ADCs, BITEs, CAR-T therapies and CAR-NK therapies [102,184,228,269,270,271]. In 2021, the ADC mAb belantamab mafodotin (GSK2857916) was the first anti-BCMA therapy to obtain approval in an indication of R/R MM (Table 2) [272]. Belantamab mafodotin is a humanized anti-BCMA IgG1 mAb linked to the antimitotic drug monomethyl auristatin F [267]. Its efficacy is being evaluated in three clinical trials (Table 4) [273]. MEDI2228 is another ADC, in which an anti-BCMA mAb is covalently linked to the pyrrolobenzodiazepine dimer tesirine. This toxin induces DNA crosslinking and the DNA damage response [274]. The results of a recent phase I study (NCT03489525) [275] demonstrated that MEDI2228 had much the same level of efficacy as belantamab mafodotin [276]. The ADC AMG224 is a BCMA-targeting IgG1 mAb coupled to the tubulin inhibitor mertansine. The results of the first phase I dose-expansion and -escalation study in R/R MM (NCT02561962, 2015–2023) evidenced significant thrombocytopenia [277]. Two BiTE anti-BCMA/CD3 agents (teclistamab and elranatamab) are currently being evaluated in as monotherapies and in combination with other treatments (Table 4) [278]. The initial results for the anti-BCMA CAR-T therapy idecabtagene vicleucel showed clinical activity and the expected adverse effects in patients with R/R (Table 4) [279]. Accordingly, the anti-BCMA CAR-T therapy ciltacabtagene autoleucel was associated with significantly better outcomes than conventional therapies in patients with R/R MM (NCT033548207) [280]. In an early phase I study, the first dual-target anti-BCMA/CD38 CAR-T (BM38) gave a high response rate, a low recurrence rate, and a manageable cytokine release syndrome in patients with R/R MM (Table 4) [281]. A now-recruiting phase I/II study is set to evaluate the safety and antitumor efficacy of BCMA-CAR-NK92 cells in patients with R/R MM (Table 4). Three phase I trials are respectively evaluating adverse events associated with the anti-BAFF-R mAb lanalumab in patients with CLL, and another trial is evaluating anti-BAFF-R-CAR-T cells in B-ALL and R/R MCL (Table 4).

3.2. Receptor Tyrosine Kinase-like Orphan Receptor (ROR1)

Aberrant expression of ROR1 is seen in CLL/SLL, MCL, and MM [47]. In CLL, ROR1 expression is associated with relatively short treatment-free and overall survival times [297,298]. As a receptor for Wnt5a [298], ROR1 can stimulate ROR1-dependent CLL cell activation of Rho-GTPases [299]. High expression levels of ROR1 and strong ROR1 signaling are associated with venetoclax resistance in CLL [300]. A phase I/II study in newly treated or R/R CLL patients showed that the mAb anti-ROR1 cirmtuzumab (recently renamed as zilovertamab) was safe and efficacious in inhibiting tumor cell ROR1 signaling in the context of R/R disease (NCT02860676, NCT02222688) [301]. Since then, several clinical trials of a combination of zilovertamab with venetoclax or ibrutunib have been initiated in patients with CLL/SLL or MCL (Table 4).

3.3. Program Cell Death 1 (PD1) and PD-Ligand 1 (PD-L1)

The immunosuppressive costimulatory signal receptor PD-1 is expressed on activated T and B cells [302,303]. PD-L1 is strongly expressed by various solid tumors, B tumor cells (including DLBCL and MM cells), and BM stromal cells [303,304]. As the tumor develops, both PD-1 and PD-L1 cause tumor immune escape–mainly by inhibiting the activity of tumor-infiltrating lymphocytes [303,304]. Several mAbs against PD1 (nivolumab, pembrolizumab, and cemiplimab) or PD-L1 (e.g., atezolizumab and durvalumab) have been approved for the treatment of various solid tumors [305,306]. Anti-PD-1 mAbs promote tumor cell apoptosis by binding to T-cell PD-1 receptors and disrupting the interaction with PD-L1 molecules on tumor cells [302]. However, no significant objective responses were observed in two phase I/Ib trials of nivolumab as a monotherapy or a combination therapy in patients with R/R MM [307,308]. Pembrolizumab exhibits selective efficacy in patients with B-NHL and CLL [283], and is currently being tested in combination with ibrutinib or fludarabine (Table 4). In a phase I/II study, a pembrolizumab + blinatumomab combination had a good safety profile (manageable adverse events) in R/R B-ALL (Table 4) [284]. However, a phase III study of a combination of pembrolizumab with pomalidomide and dexamethasone in R/R MM (NCT02576977) found a unfavorable risk-benefit ratio and was terminated [170,309]. Three clinical studies are evaluating the safety and tolerability of cemiplimab in combination with odronextamab (an anti-CD20/CD3 BiTE) or with isatuximab in patients with B-NHL (including R/R MM and DLBCL) (Table 4) [285]. A phase I study of atezolizumab alone or combined with axicabtagene ciloleucel showed a manageable safety profile in patients with R/R DLBCL and thus supported a move into phase II (Table 4) [286]. Durvalumab as a monotherapy or a combination therapy with tremelimumab (an anti-CTLA-4 mAb) or danvatirsen (AZD9150, an antisense oligonucleotide that inhibits STAT3) was well tolerated but showed limited efficacy in R/R DLBCL or FL (NCT02733042) [310,311]. The combination of durvalumab with JCAR014 (CD19/4-1BB CAR-T cells) for the treatment of patients with aggressive B-cell NHL appeared safe; although complete responses were observed in patients at initial restaging after JCAR014 infusion, and in patients continuing durvalumab therapy after initially failing to achieve complete remission (NCT02706405), the study was terminated in 2021 due to slow accrual [312].

3.4. C-X-C Chemokine Receptor Type 4 (CXCR4)

CXCR4 is the natural chemokine receptor specific for CXCL12, also termed stromal cell-derived factor-1 (SDF-1), which is released by the stromal cells residing in the thymus and in the bone marrow [313]. The overexpression of CXCR4 in solid and hematological tumors is related to disease progression [41]. In hematological malignancies, high expression of CXCR4 is observed in B cell malignancies (Table 1) [313]. Highly recurrent somatic mutations in CXCR4 gene are found in up to 40% of patients with WM [313]. Alterations in CXCR4 are associated with activation of PI3K and JAK/STAT signaling processes, which promote drug resistance by impacting BTK-inhibitor response [313]. Thus, WM patients with mutated CXCR4 are less likely to respond to ibrutinib or ibrutinib/rituximab combination therapy than WM patients with CXCR4 wild-type disease [287]. Ulocuplumab (BMS-936564) is a fully human IgG4 anti-CXCR4 mAb [314] that exhibits antitumor activity in a WM xenograft model [315]. A phase Ib/II trial showed that ulocuplumab is safe and leads to a high response rate in combination with lenalidomide and dexamethasone in patients with R/R MM (NCT01359657) [316]. An ongoing phase I/II clinical trial evaluating the combination of ibrutinib and ulocuplumab, in patients with WM, already shows that ulocuplumab dose-escalation did not impact adverse events (Table 4) [287]. Interestingly, an ongoing multicenter phase I trial is assessing the safety and tolerability of mavorixafor (AMD070, an orally bioavailable CXCR4 allosteric antagonist) in combination with ibrutinib in WM patients with CXCR4 mutation (Table 4).

3.5. CS1/Signaling Lymphocyte Activation Molecule Family 7 (SLAMF7)

CS1/SLAMF7 is overexpressed in MM cells, NK cells, plasma cells, and (to a lesser extent) some subsets of B and T cells [317]. Elotuzumab is a humanized IgG1 Ab that directly activates NK cells through the CS1 pathway and Fc receptors, thus facilitating NK-mediated ADCC [317]. In 2015, the US FDA approved elotuzumab in combination with lenalidomide and dexamethasone for the treatment of patients with MM who received one to three prior therapies, with the following warnings and precautions: infusion reactions, infections, second primary malignancies, hepatotoxicity, and interference with the determination of complete response [318]. Two phase I clinical studies are evaluating two different CS1-CAR-T constructs in patients with R/R MM (Table 4).

3.6. Other TAAS

3.6.1. CD13

The cell surface protease aminopeptidase N (APN/CD13) is overexpressed in various tumors including hematological diseases [319,320]. Based on its ability to bind to the Asn-Gly-Arg (NGR) motif, CD13 has proven to be a key for targeted delivery of NGR-bound chemotherapeutic drugs to both CD13+ tumor cells and CD13+ tumor-associated endothelium [320]. Among drugs coupled to the NGR motif, is the anti-angiogenic truncated tissue factor (tTF) that targets CD13+ tumor-associated vasculature [320]. A recent phase I trial with NGR-tTF was initiated for patients with CD13+ R/R MM (NCT02902237, completed in 2020) indicating that NGR-tTF is safely applicable with this regimen; imaging showed selective reduction of tumor blood flow and intratumoral hemorrhage and necrosis [321]. This innovative approach deserves to be pursued. A second therapeutic strategy exploited the enzymatic activity of CD13 for the activation of the alkylating prodrug melflufen (J1; dipeptide consisting of melphalan and p-fluoro-L-phenylalanine) into an active cytotoxic drug, melphalan [322]. In phase I/II studies, melflufen plus dexamethasone demonstrated encouraging clinical activity and a manageable safety profile in heavily pretreated patients with R/R MM (NCT02963493, completed in 2021) [323]. A randomized phase III study is evaluating the efficacy and safety of melflufen/dexamethasone vs. pomalidomide/dexamethasone (Table 4) [288]; melflufen plus dexamethasone shows clinically meaningful efficacy and a manageable safety profile in patients with heavily pretreated R/R MM, including those with triple-class-refractory and extramedullary disease (Table 4) [288].

3.6.2. CD16

NK cells express a CD16 (FcγRIII) V158 variant, which can exert ADCC by binding to the Fc fragment of Ig present on tumor cells [223]. Based on it, one approach was the engineering of T cells expressing CD16 CARs so that they are capable of mediating ADCC [324]. The constructs included the high-affinity CD16 V158 variant, CD8α hinge, and transmembrane domains, along with signaling domains from CD3ζ and CD28 or 4-1BB signaling domains [191,223]. Two completed first-in-human pilot studies already showed the efficacy (safety and antitumor activity) of infusing autologous T-cells engineered to express CD16V-41BB-CD3 ζ, in combination with rituximab, in patients with CLL and B-NHL (NCT02315118), and R/R B-NHL (NCT02776813) [325].

3.6.3. CD30

This antigen belongs to TNF receptor superfamily, and CD30 signaling is involved in regulation of cytokine secretion, cell proliferation and survival [326]. CD30 is highly expressed in most B-NHL [326,327,328]. Brentuximab vedotin, an ADC comprising a human anti-CD30 chimeric Ab covalently bound to the microtubule-disrupting agent monomethyl auristatin E, was approved in R/R B-NHL after autologous stem cell transplantation and anaplastic large-cell lymphoma [326]. One phase II trial is now evaluating brentuximab vedotin in combination with three chemotherapy drugs (doxorubicin, vinblastine, and dacarbazine) for B-NHL (Table 4). Anti-CD30 CAR-T cells therapies have demonstrated significant clinical responses in early clinical trials of patients with B-NHL (Table 4) [290,291]. There are other ongoing clinical trials with different CD30 CAR-T cell constructs in R/R lymphomas addressing ways to improve outcome (Table 4) (reviewed in [329,330]).

3.6.4. CD37

CD37 is expressed almost exclusively on hematopoietic cells with high expression on mature B-cells, including their malignant counterparts [331]. A large number of CD37 targeting agents have been developed including mAbs, ARCs, ADCs, and CAR-T cells [332,333,334,335]. Among them, the anti-CD37 BI856826, initially reported as a valid therapeutic target in B-NHL (NCT01403948) [336] and CLL (NCT02759016) [334], did not undergo further clinical development upon the decision of sponsors to discontinue the trials. A phase I/II study is evaluating the safety and efficacy of the ARC 177Lutetium-lilotomab satetraxetan (betalutin) for treatment of relapsed B-NHL (Table 4) [289]. In contrast to other newly developed CD37 targeting agents, the duoHexaBody-CD37 (GEN3009, a novel biparatopic CD37-bispecific IgG1 with an E430G hexamerization- enhancing single point mutation in the Fc-domain), mediates potent CDC in addition to ADCC and cellular phagocytosis [131]. A first-in-human clinical trial is evaluating GEN3009 in patients with R/R B-NHL (Table 4). Naratuximab emtansine (IMGN529, Debio1562) is an ADC anti-CD37 mAb conjugated to maytansine-derived microtubule disruptor; it was investigated alone in CLL and R/R B-NHL (NCT01534715) [337], and in combination with rituximab in R/R DLBCL, FL and MCL (NCT02564744), where the treatment was well-tolerated and resulted in complete response rates and a manageable safety profile [338]. The second ADC, AGS67E, is an anti-CD37 mAb conjugated to monomethyl auristatin E, that showed a favorable safety profile in a phase I clinical trial in patients with CLL and R/R B-NHL (NCT02175433) [339]. Extended studies are awaited.

3.6.5. CD46

CD46 (also referred to as membrane cofactor protein, MCP) is coexpressed as four predominant isoforms on almost all cell types, and is overexpressed on a variety of human tumor cells [340]. Clinical and experimental data support an association between increased CD46 expression, malignant transformation, and metastasizing potential [340]. More recently, CD46 has been reported as a driver of metabolic processes [340]. The ADC anti-CD46-monomethyl auristatin E (FOR46) demonstrated an acceptable toxicity profile in a phase I study to treat patients with R/R MM (NCT03650491) [341].

3.6.6. CD47

This ubiquitous antigen belongs to the immunoglobulin superfamily [342], and exhibits various cellular functions with multiple binding partners, such as inhibition of phagocytosis through an interaction with signal-regulating protein alpha (SIRPα) on the surface of phagocytic cells [343]. Many clinical trials have started to evaluate numerous anti-CD47 mAbs (and derivatives) in the treatment of solid tumors and hematological neoplasms including B cell diseases [343,344]. Monotherapy phase I clinical trials of anti-CD47 Abs often yielded limited signs of efficacy compared with combination strategies [343]. Among them, the humanized anti-CD47 mAb magrolimab selectively binds to CD47 expressed on tumor cells and blocks the interaction of CD47 with SIRPα, thus allowing the activation of macrophages, resulting in specific tumor cell phagocytosis [345]. Furthermore, blocking CD47 signaling activates an antitumor T-lymphocyte immune response and T-mediated cell killing [345]. Of interest, the phase II study (NCT04778397) enrolling patients with AML or myelodysplastic syndrome who received magrolimab in combination with azacytidine, was paused in 2022 following suspicions of safety issues [346]. Thus, particular attention should be paid to magrolimab, which is currently tested in combination with standard therapies in several trials in B-NHL (Table 4). A second anti-CD47 (JMT601/CPO107) is a bispecific fusion protein in which the CD47 binding fragment SIRPα is covalently fused to ofatumumab (an anti-CD20); it thus targets both CD20 and CD47 on tumor cells and leads to ADCC and CDC [344]. A phase I/II multicenter clinical trial in CD20+ B-NHL patients is starting (Table 4).

3.6.7. CD56

This antigen, primarily expressed in cells of neuroendocrine origin as well as in NKs and some T cell subtypes, is overexpressed in several solid and hematologic malignancies, including MM [347]. The ADC lorvotuzumab mertansine (IMGN901) combines the anti-CD56 Ab lorvotuzumab covalently bound to the cytotoxic maytansinoid effector. A phase I trial of lorvotuzumab mertansine provided evidence of safety and signals of clinical activity for this agent in R/R MM (NCT00346255) [348], warranting its further clinical development as part of combination regimens for the management of MM.

3.6.8. CD74

Otherwise known as HLA-DR-associated invariant chain, CD74 functions as an MHC class II chaperone and as a high affinity receptor for the pro-inflammatory cytokine macrophage migration inhibitory factor [349]. CD74 is overexpressed in some B-NHL [349]. A phase I study with the anti-CD74 mAb milatuzumab [350] showed no objective responses in R/R MM (NCT00421525) [351]. ASTRO-001 is a fully human ADC anti-CD74 incorporating maytansinoid [352]; a phase I study of ASTRO-001 is recruiting patients with advanced B cell malignancies including DLBCL, FL and MM (Table 4) [294].

3.6.9. CD79B

CD79 is the BCR’s signaling component (Figure 1b) and is composed of a heterodimer of CD79A and CD79B, both of which induce internalization of the BCR [353]. CD79 is strongly expressed in most types of B-NHL and CLL but weakly expressed on normal cells (with restriction to pre-B and mature B cells) [353]. Polatuzumab vedotin is an ADC humanized anti-CD79B Ab conjugated to monomethyl auristatin E, that showed significant clinical activity in phase I trials to treat patients with FL and DLBCL [353]. In 2019, it was FDA-approved in combination with bendamustine and rituximab for patients with R/R DLBCL after at least two prior therapies [353]. Two phase I trials are evaluating polatuzumab vedotin in combination with several conventional chemotherapies for untreated or R/R patients with DLBCL and other B-NHL (Table 4).

3.6.10. CD156/a Disintegrin and Metalloprotease 17 (ADAM17) (Also Referred to as TNF-α Converting Enzyme/TACE)

The cell surface protease ADAM17 is overexpressed in certains solid and hematological tumors [354], and the development of a large variety of inhibitors against the enzymatic activity of ADAM17 has made it into clinical evaluation in cancers [320]. Among these ADAM17 inhibitors, aderbasib (INCB7839) [355] entered a phase I/II clinical trial, to be used along with rituximab as consolidation therapy after an autologous hematopoietic cell transplant for patients with DLBCL (NCT02141451) (Table 4). The short-term results suggest applicability as relapse preventing therapy for DLBCL [356].

3.6.11. CD307/Fc Receptor-like 5 (FcRL5)

CD307/FcRL5, specifically expressed within the B cell lineage [357], belongs to transmembrane FcRL family proteins with sequence homology to classical Fc receptors, which can promote B cell proliferation and activation [317]. Present on malignant cells from patients with MM, CLL and MCL, CD307 may be therefore a potential therapeutic target for the treatment of these diseases [358]. Cevostomab (BFCR4350A) is a BiTE anti-FcRL5/CD3 Ab being evaluated in an ongoing phase I, dose escalation and expansion trial (Table 4) [295].

3.6.12. G-Protein Coupled Receptor Family C Group 5 Member D (GPRC5D)

GPRC5D is ubiquitously expressed on healthy and malignant plasma cells, and high expression on MM cells associated with adverse prognosis [359]. Talquetamab is a BiTE duobody anti-GPRC5D/CD3 currently evaluated in a phase I study, in which it is administered both intravenously or subcutaneously in patients with R/R MM (Table 4); it already shows a high clinical response rate and appears well-tolerated, allowing the next exploration of dosing strategies [296].

4. Targeting Metabolic Reprogramming in B Cell Malignancies—Preclinical and Clinical Studies

4.1. Metabolic Reprogramming in B Cell Malignancies

4.1.1. The Metabolism in Cancer Cells, the Warburg Theory

The main characteristics of tumor cells (e.g., evading cell death, insensitivity to anti-growth signals, limitless replicative potential, tissue invasion and metastasis, sustained angiogenesis) were defined two decades ago as “the hallmarks of cancer” [360]. Ten years later, the same authors updated this model of acquired capabilities of cancer by including two extra features that emerged as crucial factors for cancer growth and progression, e.g., metabolic reprogramming and immune evasion [361]. When considering the current treatments of B cell malignancies, both hallmarks are very promising and intensively studied sources of inspiration for the development of new therapeutic strategies. Besides the excitement for immunotherapy with a tremendous number of engineered CAR-T/CAR-NK cells strategies (Section 3.4), the discovery of metabolic vulnerabilities in tumor cells is also likely to profoundly impact the development of new therapeutic options [362].
To compensate for its elevated bioenergetic needs associated to increased proliferation and biosynthesis, tumor cells must adapt and reprogram their metabolism to increase nutrients import. In particular, a dramatic increase in glucose uptake and a shift toward its oxidation through aerobic glycolysis (referred to as the “Warburg effect”) is observed in most tumor cells (Figure 3) [362]. Glucose is first converted into pyruvate through 10 enzymatic reactions, the first of which is catalyzed by hexokinase 2 (HK2) (Figure 3). Pyruvate, the final product of glycolysis, is further transformed into lactate by the lactate dehydrogenase-A (LDHA) (Figure 3). Another major alternative to produce energetic ATP occurs through mitochondria, where the oxidation of various nutrients (including amino acids such as glutamine, or lipids such as free fatty acids/FFA) can feed the tricarboxylic (TCA) cycle (Figure 3). The TCA can then activate mitochondrial respiration through the electron transport chain, which promotes oxidative phosphorylation (OXPHOS) and silences the Warburg effect and the shift to glycolysis (Figure 3).

4.1.2. Specific Metabolic Features of B Cell Malignancies

While the Warburg effect describes the typical metabolic response in aggressive B-NHL [363], other B cell malignancies favor mitochondrial respiration, such as CLL.
Glycolysis: the expression of the rate limiting enzyme (HK2, an indicator of glycolytic activity) is often upregulated in B-NHL [364,365]. No change in glycolysis is observed in CLL, although an increase in glycolytic capacity and reserve appears linked to drug resistance [366]. As a consequence, the blockade of the GLUT4 glucose transporter by the HIV protease inhibitor ritonavir leads to in vitro cell death in CLL [367] and potentiates the efficacy of metformin in MM cell lines and xenograft models [368]. A recent clinical trial demonstrated the safety, tolerability and feasibility of metformin + ritonavir combination therapy in patients with R/R MM or CLL (NCT02948283) [369]. Furthermore, the expression of lactate dehydrogenase A (LDHA) is upregulated in NHL [370] including MM [210]. Because of the involvement of glycolysis in B-NHL progression, the targeting of lactate trafficking through the blockade of monocarboxylate transporter 1 (MCT-1) by AZD3965 shows very promising results in NHL [371], including DLBCL [372] A phase I clinical trial of AZD3965 combined to rituximab in DLBCL has recently been completed (NCT01791595) [373]. Independently of glycolysis, one regulatory mechanism has been described in B-cell malignancies by which glucose is redirected toward glycolysis or the pentose phosphate pathway (PPP), offering new options with the LB-100 inhibitor [374].
Mitochondrial OxPhos: an increase in OxPhos has been observed in MCL [375], MM [376] and CLL [377,378]. The increase in OXPHOS in CLL leads to increase in the oxidative stress [379], and forcing mitochondrial respiration through PI3K/AKT hyperactivation leads to toxic accumulation of ROS, resulting in CLL cell death [380]. In DLBCL, the combination of AZD3965 with the mitochondrial respiration inhibitor IACS-010759 in B cell lines and xenografts suggests a clear clinical advantage compared with monotherapy [381]. The novel mitochondrial disrupter CPI-613 (devimistat, a lipoate analog) inhibits the mitochondrial enzymes pyruvate dehydrogenase and α-ketoglutarate dehydrogenase in the TCA cycle [382,383]. A phase II study with CPI-613 is now recruiting patients with R/R NHL, including those with high-risk translocations of Myc, BCL2 and/or BCL6 (NCT03793140).
Lipids: beside carbohydrates metabolism, the metabolism of lipids is also affected in B-cell malignancies [264]. Lipid metabolism is upregulated in BL [384]. In CLL, high expression of lipoprotein lipase LPL is associated with a shorter treatment-free survival and a trend of a shorter median overall survival [385]. Its overexpression causes an increase in the β-oxidation of FFAs [386], while its inhibition by orlistat induces apoptosis of CLL cells [387]. Orlistat also induces cell death in MCL by inhibiting fatty acid synthase (FASN) [388] while pharmacological or genetic inhibition of FASN in DLBCL also induces apoptosis [389]. The antigen CD36 that facilitates FFA uptake by cells, is upregulated in CLL, and its inhibition by anti-CD36 neutralizing Abs or sulfosuccinimidyl oleate (SSO) leads to apoptosis [390]. Similarly, the blockade of FFAs import into mitochondria via their β-oxidation by perhexiline (inhibitor of carnitine palmitoyltransferases (CPT), essential for transporting long chain fatty acid into mitochondria) [391] or the irreversible inhibitor of CPT-1, etomoxir [377], induces CLL cell death.
Amino acids: amino acids play a crucial role in the progression of B-cell malignancies. In peculiar, addiction of tumor cells to glutamine is observed in many tumors including MM [392] and CLL with 11q deletion [393], and in the Burkitt lymphoma cell line P493 [394]. The glutaminase inhibitor CB-839 (telaglenastat) has been tested in phase I clinical trial in MM and WM, but the results are not yet available (NCT02071888).

4.1.3. Involvement of the Tumor Microenvironment in the Adaptative Metabolism

Often, tumor cells depend on microenvironmental non-malignant cells for survival. Indeed, while some intrinsic common metabolic characteristics can be observed in B cell malignancies, metabolism can be modified by the contact of tumor cells with the adjacent tumor microenvironment (TME) [395,396]. For instance, interaction of tumor CLL cells with surrounding bone marrow (BM) stromal cells is shown to induce a shift toward glycolysis [379,397], that is in part mediated by Notch/c-Myc signaling [379]. Protection by the TME involves the transcriptional regulation of genes involved in redox homeostasis [398]. Furthermore, the protection of CLL cells from oxidative stress is also handled through a metabolic collaboration with stromal cells which transform cystine to cysteine, and cysteine transport to CLL cells allows the synthesis of GSH to promote cell survival and drug resistance [399]. As recently reviewed by Nie et al. [378], the agents targeting metabolic enzymes or signal pathways activated in CLL by lymph nodes (LN) microenvironment (converging to glucose, purine and glutamine metabolism) may effectively impede the progression of CLL [400]; these data provide further insight into the crucial metabolic changes driven by the CLL LN microenvironment and the prominent use of amino acids as fuel for the TCA cycle, in view of new therapeutic vulnerabilities. A high mitochondrial respiration in MM and B-ALL cells appears to be the consequence of the direct transfer of mitochondria from stromal to tumor cells [401,402]; this phenomenon in MM cells is driven by CD38 [402]. Tumor-associated adipocytes support MM cells through various mechanisms, including metabolic reprogramming of MM cells which involves MM cell–induced lipolysis in BM adipocytes, upregulation of FA transporters 1 and 4 on MM cells which then mediate the uptake of secreted free FAs, leading to enhanced MM proliferation [403].
In contrast, tumor-associated stromal cells can metabolically stimulate the growth and metastasis of cancer by producing a “reverse Warburg effect” [404,405]. Thus, stromal cells shift from glycolysis to mitochondrial respiration in cases of B-NHL [406,407]; in particular, the lactate trafficking that promotes tumor growth in NHL is caused by the upregulation of MCT4 in tumor-associated macrophages and the concomitant upregulation of MCT-1 in tumor cells [371,408]. The blockade of lactate by AZD3965 leads to NHL cell apoptosis [371,408].
In conclusion, these examples of metabolic vulnerabilities open up new opportunities for the treatment of B cell malignancies by cutting off the tumor’s supplies of energy substrates.

4.2. Targeting Metabolism in B Cell Malignancies: Clinical Perspectives

4.2.1. Metabolic Features as Biomarkers of B Cell Malignancies

Metabolomic approaches on plasma of patients with B cell malignancies may allow to identify metabolic biomarkers for early diagnosis and prediction, prognosis and treatment [409]. Although many metabolic biomarkers have already shown their clinical validity, many others have not yet been subjected to extensive testing to demonstrate their clinical usefulness in B-cell malignancies. For instance, three metabolic subtypes have been identified in nine B-NHL model cell lines according to their preferential use of glucose, glutamine or both as a source of energy [410]. As an attempt to perform a classification of B-NHL according to their specific metabolic profile, a pilot study was performed on the plasma samples of 66 NHL patients, compared with 96 matching controls [411]. A significant difference was observed between the metabolites produced by DLBCL, CLL, MM, NHL patients’ plasma (but not FL) and their respective control population, allowing to discriminate these patients and healthy controls [411]. Although the NHL cohort size was rather small, the results already suggest a classification of NHL subtypes based on plasma metabolites [411]. In MM, the serum metabolic profiling shows a specific signature after complete remission [412]. A similar analysis of serum from 47 childhood ALL patients lead to the identification of 30 potential biomarkers (17 detected in positive mode and 13 in negative mode) [413]; in particular, deregulated glycerophospholipid metabolism in ALL patients appeared to represent an underlying metabolic pathway associated with disease progression [413]; this non-invasive diagnosis approach may especially be relevant as diagnosing ALL requires BM puncture procedure.
In CLL, IGHV mutational status correlates with disease-related metabolic profiles [366,414]. Several key glycolytic genes (PFKP, PGAM1 and PGK1) were found to be down-regulated in samples harboring mutated immunoglobulin variable heavy-chain. Furthermore, 8q24 copy number gains, 8p12 deletions, 13q14 deletions and ATM mutations were identified as determinants of cellular respiration [366]. Such heterogeneity in the energy metabolism of CLL cells could be therapeutically exploited in the selection of therapeutic strategies. In DLBCL, a genome-wide array study identified three clusters including one defined by increased OXPHOS and expression of genes involved in mitochondrial oxidative phosphorylation (OxPhos-DLBCL). OxPhos-DLBCL also overexpresses proteasome and apoptosis regulators, suggesting a better response to drugs targeting mitochondrial function, proteasome and BCL2 family [415]. A further characterization of these three DLBCL subtypes detailed the metabolic signature of each of the subtypes [415]. The blockade of FAO and GSH synthesis is selectively toxic to OxPhos-DLBCL [416]. Conversely, the absence of functional BCR signaling in OxPhos-DLBCL renders it insensitive to BCR inhibition contrary to non-OxPhos-DLBCL [417], resulting in enhanced glycolysis [416]. More recently, a very comprehensive study identified the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as a biomarker for distinguishing between OxPhos-DLBCL (GAPDHlow) and non-OxPhos-DLBCL (GAPDHhigh) and a clinical marker for predicting overall survival in patients treated with R-CHOP [418].

4.2.2. New Strategies for Targeting Metabolism in B Cell Malignancies and Optimizing Current Treatments

While metabolic inhibitors have been used for decades for the treatment of metabolic disorders, the acknowledgement of metabolic reprogramming as a crucial feature of cancer biology has recently started to open new perspective for cancer treatment, with the repositioning of commercial metabolic drugs, or the design of new inhibitors evaluated in clinical trials [419,420]. As the inhibition of major metabolic pathways may have a dramatic impact on both tumor and normal cells, current experience shows that the best potential of metabolic inhibitors to treat tumor, comes from their combination with classically used drugs, as a way to overcome drug resistance [421]. Importantly, while tumor progression correlates to a change in metabolic profile, acquisition of resistance to drug is, most of the time, also linked to a change in bioenergetics. Thus, resistance to chemo-immunotherapy in CLL correlates to the activities of glycolytic enzymes LDHA and pyruvate kinase M2 (PKM2), leading to the development of a metabolic score as a diagnostic tool in CLL [422]. A larger CLL screening study revealed that the metabolic state of leukemic cells is associated with drug sensitivity; in particular, higher glycolytic activity is linked to increased resistance towards drugs (including venetoclax, ibrutinib, R-CHOP, rituximab, proteasome inhibitors) thus shedding some light on metabolic vulnerabilities displayed by resistant cells [366].
Venetoclax: two main metabolic patterns emerge in venetoclax resistant B-NHL and CLL. First, mitochondrial OxPhos is increased in response to venetoclax in B-NHL, rendering them more sensitive to respiration inhibitors such as antimycin, oligomycin, IACS-010759, metformin or berberine [423,424,425,426]. Second, inhibition of LN-derived glutamine import with V9302 attenuates LN-induced resistance of CLL cells to venetoclax [400]; a higher glycolytic activity is also part of the resistant phenotype [366].
Ibrutinib: ibrutinib treatment which blocks BCR signaling also interferes with metabolic pathways in CLL, decreasing mitochondrial oxidative respiration, glycolysis and glutamine metabolism [400,427,428] An increase in glutamine uptake is also observed in ibrutinib-resistant CLL cells, and etomoxir treatment (that blocks the mitochondrial import of FAs and their βeta-oxidation), has a cytotoxic effect on ibrutinib resistant CLL cells [377]. In contrast, ibrutinib-resistant MCL cells display increased OxPhos, and combining ibrutinib to IACS-010759 blocks tumor growth [375]. Ibrutinib resistance also correlates to higher glutaminolysis, that renders glutaminase inhibitor CB-839 efficient at killing ibrutinib-resistant MCL cells [429].
R-CHOP: Low GAPDH expression level correlates to DLBCL patients’ resistance to R-CHOP treatment [418,430], and their sensitivity to metabolic inhibitors targeting mammalian target of rapamycin complex 1 (mTORC1) signaling, OxPhos and glutaminolysis [418]. In a preliminary clinical trial, four DLBCL patients with GAPDHlow tumors received a combination therapy involving kidrolase (asparaginase), metformin, and temsirolimus (mTOR inhibitor), tumor regression was reported in three out of four patients for the first two weeks of treatment, but relapsed after longer treatment, with a median response duration of six months [430]. This preliminary study suggests that combination of metabolic inhibitors may offer a treatment strategy for metabolically active cancers [430].
Rituximab: acquisition of resistance to rituximab appears related to an increase in glycolysis in BL and DLBCL cell lines, and downregulation of HK2 by 2-Deoxy-D-glucose (2DG) glucose competitive inhibitor, metformin, idelalisib or temsirolimus overcomes resistance and enhances the cytotoxic effect of rituximab [431]. Similarly, the blockade of lactate transporter MCT1 by AZD3965 combined to rituximab leads to tumor regression in a BL mouse xenograft [432] as well as in patients with DLBCL (phase I NCT01791595, completed in 2020) [373]. An enhancement of rituximab response is observed in DLBCL when combined with metformin [433] or pyruvate dehydrogenase kinase 4 (PDK4) inhibitor dichloroacetate (DCA) [434]. Both temsirolimus and everolimus are already FDA-approved for the combined treatment with rituximab of R/R MCL, DLBCL and FL [435].
Proteasome inhibitors: in MM cell lines and a MM mouse xenograft, the response to proteasome inhibitors is increased when combined to the glutaminase inhibitor CB-839 [436] or to DCA [437].

4.2.3. Metabolism as a Tool to Boost Immunotherapy

In the last years, the development of immunotherapy has been quite impressive for the treatment of various tumors including B-cell malignancies. Despite some very promising results, this new therapy is facing long term efficacy issues, due to the difficulty to maintain the proliferation of immune cells in an immunosuppressive TME. One major limitation is the availability of nutriments in the TME, for which immune and tumor cells compete; the high avidity of tumor cells for energy substrates, especially glucose, often results in exhaustion of immune cells, ending with tumor immune evasion [438]. Moreover, the accumulation of immune suppressive metabolites in the TME has negative effects on the immune system, in particular lactate production by tumor cells can inhibit the activity of B and T cells [439]. Complementing immunotherapy with molecules that modulate metabolism therefore emerges as a solution of choice to optimize immunotherapies, including chimeric antigen receptor (CAR) T cells [440,441]. For instance, glutaminolysis in DLBCL interferes with immunotherapy by regulating positively PD-L1 expression [442]. Two phase I clinical trials in B-NHL evaluated the combinations of pembrolizumab with troriluzole (BHV-4157), an inhibitor of glutamate (NCT03229278, completed in 2020), or ciforadenant, an antagonist of the adenosine A2A receptor (NCT03454451, 2018–2025). These innovative approaches deserve to be pursued.

5. Concluding Remarks and Perspectives

Today, a key challenge in using FDA-approved drugs in the current treatment of B cell malignancies (Figure 2a) is that these therapies must simultaneously eliminate tumor cells and preserve/reestablish normal hematopoiesis. However, the effectiveness of these anticancer therapies is sometimes limited by primary and acquired resistance [1]. Drug-mediated T cell exhaustion may lead to the possibility of immune escape of hematological malignancies [1]. Residual disease and/or resistance in patients treated with Bcl-2/BTK/PI3K inhibitors (venetoclax, ibritunib, idelalisib, etc.) is associated with genomic resistance (mutations, activation, instability, p53 aberrations.) as well as non-genomic, acquired resistance through (re)activation of signaling survival pathways (PI3K, NF-κB, MYC, etc.), cancer stem cells, and the tumor microenvironment [60,443,444,445,446,447]. The main adverse effects of mAbs therapies include cytokine release syndrome, neurotoxicity, and on-target/off-tumor toxicity resulting from a direct attack on normal tissues that have shared expression of the target antigen [223,448]. To improve clinical outcomes (residual and progressive disease) and immune function in patients with B cell malignancies, while evading the emergence of drug resistance, newer drugs that inhibit the same targets as the current treatments are in continuous clinical development. For instance, ongoing trials focus on overcoming venetoclax resistance by targeting the BCL2 family members Mcl-1 and Bcl-xL, and evaluate more highly selective BTK and PI3Kδ inhibitors with fewer off-target effects. These second-generation drugs, although more potent, are not likely to be used only as single agents. Drug combinations can favor compounds with synergistic antitumor effects and ensure manageable toxicity. Thus, the combinations of BTK inhibitors with BCL2 inhibitors and/or rituximab (anti-CD20 mAb), or PI3K inhibitors with BCL2 inhibitors, are of interest.
Importantly, this review outlines the increasing importance of therapeutic mAbs (mono-/bi-specific, ADC, BiTE, CAR-T cells, Bi-CAR-T cells, CAR-NK cells) as the predominant treatment modality for B cell malignancies. To face the resistance mechanism, BiTEs and CAR-T cells redirect T cells to target antigen-expressing tumors; novel BiTEs and CAR-T cells with various costimulatory signals and delivery systems, as well as Bi-CAR-T cells (anti-CD19/CD20 CAR-T, anti-CD19/CD22 CAR-T, and anti-CD20/CD22 CAR-T) may therefore represent an effective solution to the challenge of antigen escape in immunotherapy (Table 3). As already planned for the treatment of solid tumors [449], BiTEs and CAR-T cells could be combined with standard chemotherapy and/or targeted therapies of B cell malignancies to reduce the tumor burden and/or modulate the immune response. Recently, CAR-T cells and BiTEs have been engineered into a single immunotherapy platform (BiTE-secreting CAR-T) for T-cell-directed therapy of solid tumors [450,451]. BiTEs secreted by CAR-T cells exhibit potent antitumor activity in vitro and in vivo with significant sensitivity and specificity, demonstrating a promising strategy in solid tumor therapy [450,451]. This approach could have therapeutic application in hematological malignancies. Lastly, compared with CAR-T cells, CAR-NK cell constructs offer advantages, including better safety, and multiple mechanisms for activating cytotoxic activity [452,453,454]. For therapy of B cell malignancies, equipping anti-CD19 CAR-NK cells with on-board cytokines or chemokines might improve clinical efficacy by enhancing both persistence and cytotoxicity against tumor cells.
The consideration of other TAAs (including BCMA, BAFF-R, ROR1, CXCR4, CS1, FcRL5, GPRC5D, etc.) as druggable targets for B cell malignancy immunotherapy, has promoted the development of novel inhibitors, with some of them under single or combined evaluation in clinical trials for a variety of patients with B cell malignancies (Table 4). Most TAAs show their potential for ADC, BiTE, CAR-T, and CAR-NK therapies. Regarding the PD1/PD-L1 axis responsible in part for cancer immune escape, immune checkpoint blockade therapies with anti-PD1/anti-PD-L1 mAbs in combination with BTK (ibrutinib), anti-CD38 (isatuximab) or anti-CD19 CAR-T (axicabtagene ciloleucel) may optimize the landscape of B-NHL therapy.
Targeting additional intracellular signaling pathways contributing to B malignant cell dysfunction may also prove efficacious. For example, ruxolitinib, a potent JAK1/2 inhibitor, demonstrates antitumor activity as a single agent in MM (NCT03110822) [455]. Anvatirsen, an antisense oligonucleotide inhibiting STAT3, combined with durvalumab + tremelimumab is well-tolerated in patients with R/R DLBCL (NCT02807454) [311]. The efficacy and safety of a combination of panobinostat (a pan-histone deacetylase inhibitor) with bortezomib + dexamethasone is being evaluated in MM patients having received two or more lines of treatment (NCT02654990). Tazemetostat/EPZ-6348, which inhibits the histone methyl transferase EZH2, shows safety and antitumor activity in patients with DLBCL (NCT02889523) and FL (phase I/II NCT01897571) [456,457,458]. Combination therapy with temsirolimus, an inhibitor of the oncogenic kinase mTOR, and lenalidomide demonstrates encouraging activity in patients with DLBCL and FL (NCT01076543) [459]. Ponatinib is a third-generation tyrosine kinase inhibitor with a wide spectrum of kinase inhibition [460]; it targets BCR-ABL1, an abnormal tyrosine kinase that is expressed in chronic myeloid leukemia and Philadelphia chromosome-positive (Ph+) ALL; ponatinib shows clinical activity in R/R Ph+ (BCR-ABL) ALL [461], and in the first-line setting in combination with standard chemotherapy (NCT01641107, [462]; NCT02776605, [463]) or blinatumomab (NCT03263572) [464]. A Phase III study is comparing the efficacy and the safety of the first in-class selective inhibitor of nuclear export, selinexor, in combination with bortezomib + dexamethasone vs. bortezomib + dexamethasone in patients with R/R MM (NCT03110562) [465]. Pharmacological inhibition of the bromodomain and extra-terminal (BRD/BET) family proteins block downstream components of BCR signaling, downregulate Bcl-2 transcription, and suppress NF-κB signaling in CLL and B-NHL [466]; among novel single-molecules cotargeting BRD4 and other tumor targets recently developed, SRX3177 targeting CDK4/6-PI3K-BRD4 and SRX3305 targeting BTK-PI3K-BRD4, demonstrate preclinical activity against MCL [467], CLL [468], and MCL [469]. These studies underscore the potential effectiveness of these novel multi-action small molecule inhibitors, alone or combined, for potential treatment of B tumors.
Last, but not least, metabolic changes in tumor cells represent a novel opportunity for combination therapy approaches [470]. Metabolic reprogramming is linked to oncogenic transformation. One hallmark of BCL2, PI3K, BTK, and mTOR proteins and some TAAs such as CD38, concerns their participation in tumor metabolism. Activation of these oncogenic pathways makes tumors more metabolically active, and conversely, active metabolism upregulates BCL2, PI3K, BTK, and mTOR proteins, inhibiting susceptibility to cell death. In a consistent way, a few clinical trials have started to include metabolic inhibitors (targeting for instance glycolysis, OxPhos, amino acids) with venetoclax, ibrutinib and other drugs, to overcome the limitations of targeted therapeutic strategies in lymphoid malignancies. As metabolic reprogramming is closely linked to tumor B cells’ microenvironment and to immunoevasion, strategies targeting these crosstalks may also open new avenues for overcoming therapeutic resistance.
To conclude, most therapeutic drugs have underscored our advancement in the understanding of the biology of malignant B cells, and has improved outcomes for many patients. In the search for better efficacy and safety, the status of therapies in B cell malignancies is continually advancing with emerging concepts in therapy and evolving results from clinical protocols. A large variety of more effective and selective inhibitors and targeted combinations are being evaluated in these diseases, and it is expected that some of these new compounds will proceed into the clinic in the near future.

Author Contributions

M.T., D.G. and B.B. wrote the article. M.T., D.G., S.A.S. and B.B. have corrected, read and agreed to the published version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Institut National de la Santé et de la Recherche Médicale (Inserm). M.T. received PhD fellowships from SIRIC-CURAMUS (INCA-DGOS-Inserm_12560) and the Société Française d’Hématologie (SFH).

Acknowledgments

This review is dedicated to Sante Tura (1929–2021), internationally renowned hematologist, pioneer of modern Italian hematology, who launched a large program of clinical research and assistance for people with hematologic cancers.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef] [PubMed]
  2. Tessoulin, B.; Papin, A.; Gomez-Bougie, P.; Bellanger, C.; Amiot, M.; Pellat-Deceunynck, C.; Chiron, D. BCL2-Family Dysregulation in B-Cell Malignancies: From Gene Expression Regulation to a Targeted Therapy Biomarker. Front. Oncol. 2018, 8, 645. [Google Scholar] [CrossRef]
  3. Adams, C.M.; Clark-Garvey, S.; Porcu, P.; Eischen, C.M. Targeting the Bcl-2 Family in B Cell Lymphoma. Front. Oncol. 2019, 8, 636. [Google Scholar] [CrossRef] [PubMed]
  4. Kapoor, I.; Bodo, J.; Hill, B.T.; Hsi, E.D.; Almasan, A. Targeting BCL-2 in B-cell malignancies and overcoming therapeutic resistance. Cell Death Dis. 2020, 11, 941. [Google Scholar] [CrossRef] [PubMed]
  5. Morales-Martínez, M.; Vega, M.I. Roles and Regulation of BCL-xL in Hematological Malignancies. Int. J. Mol. Sci. 2022, 23, 2193. [Google Scholar] [CrossRef] [PubMed]
  6. Sulkshane, P.; Teni, T. Myeloid cell leukemia-1: A formidable barrier to anticancer therapeutics and the quest of targeting it. Explor. Target Antitumor Ther. 2022, 3, 278–296. [Google Scholar] [CrossRef] [PubMed]
  7. Roberts, A.W.; Huang, D. Targeting BCL2 With BH3 Mimetics: Basic Science and Clinical Application of Venetoclax in Chronic Lymphocytic Leukemia and Related B Cell Malignancies. Clin. Pharmacol. Ther. 2017, 101, 89–98. [Google Scholar] [CrossRef] [PubMed]
  8. Argyropoulos, K.V.; Vogel, R.; Ziegler, C.; Altan-Bonnet, G.; Velardi, E.; Calafiore, M.; Dogan, A.; Arcila, M.; Patel, M.; Knapp, K.; et al. Clonal B cells in Waldenström’s macroglobulinemia exhibit functional features of chronic active B-cell receptor signaling. Leukemia 2016, 30, 1116–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Saba, N.S.; Liu, D.; Herman, S.E.; Underbayev, C.; Tian, X.; Behrend, D.; Weniger, M.A.; Skarzynski, M.; Gyamfi, J.; Fontan, L.; et al. Pathogenic role of B-cell receptor signaling and canonical NF-κB activation in mantle cell lymphoma. Blood 2016, 128, 82–92. [Google Scholar] [CrossRef] [Green Version]
  10. Efremov, D.; Turkalj, S.; Laurenti, L. Mechanisms of B Cell Receptor Activation and Responses to B Cell Receptor Inhibitors in B Cell Malignancies. Cancers 2020, 12, 1396. [Google Scholar] [CrossRef]
  11. Paillassa, J.; Safa, F.; Troussard, X. Updates in hairy cell leukemia (HCL) and variant-type HCL (HCL-V): Rationale for targeted treatments with a focus on ibrutinib. Ther. Adv. Hematol. 2022, 13, 20406207221090886. [Google Scholar] [CrossRef] [PubMed]
  12. Profitós-Pelejà, N.; Santos, J.C.; Marín-Niebla, A.; Roué, G.; Ribeiro, M.L. Regulation of B-Cell Receptor Signaling and Its Therapeutic Relevance in Aggressive B-Cell Lymphomas. Cancers 2022, 14, 860. [Google Scholar] [CrossRef]
  13. Burger, J.A.; Wiestner, A. Targeting B cell receptor signalling in cancer: Preclinical and clinical advances. Nat. Rev. Cancer 2018, 18, 148–167. [Google Scholar] [CrossRef] [PubMed]
  14. Gambella, M.; Carlomagno, S.; Raiola, A.M.; Giannoni, L.; Ghiggi, C.; Setti, C.; Giordano, C.; Luchetti, S.; Serio, A.; Bo, A.; et al. CD19-Targeted Immunotherapies for Diffuse Large B-Cell Lymphoma. Front. Immunol. 2022, 13, 837457. [Google Scholar] [CrossRef]
  15. Pavlasova, G.; Mraz, M. The regulation and function of CD20: An “enigma” of B-cell biology and targeted therapy. Haematologica 2020, 105, 1494–1506. [Google Scholar] [CrossRef]
  16. Shah, N.N.; Sokol, L. Targeting CD22 for the Treatment of B-Cell Malignancies. Immunotargets Ther. 2021, 10, 225–236. [Google Scholar] [CrossRef] [PubMed]
  17. Horenstein, A.L.; Faini, A.C.; Morandi, F.; Bracci, C.; Lanza, F.; Giuliani, N.; Paulus, A.; Malavasi, F. The Circular Life of Human CD38: From Basic Science to Clinics and Back. Molecules 2020, 25, 4844. [Google Scholar] [CrossRef] [PubMed]
  18. Buhl, A.M.; Pleiman, C.M.; Rickert, R.C.; Cambier, J.C. Qualitative regulation of B cell antigen receptor signaling by CD19: Selective requirement for PI3-kinase activation, inositol-1,4,5-trisphosphate production and Ca2+ mobilization. J. Exp. Med. 1997, 186, 1897–1910. [Google Scholar] [CrossRef] [Green Version]
  19. Deaglio, S.; Capobianco, A.; Bergui, L.; Dürig, J.; Morabito, F.; Dührsen, U.; Malavasi, F. CD38 is a signaling molecule in B-cell chronic lymphocytic leukemia cells. Blood 2003, 102, 2146–2155. [Google Scholar] [CrossRef]
  20. Polyak, M.J.; Li, H.; Shariat, N.; Deans, J.P. CD20 homo-oligomers physically associate with the B cell antigen receptor. Dissociation upon receptor engagement and recruitment of phosphoproteins and calmodulin-binding proteins. J. Biol. Chem. 2008, 283, 18545–18552. [Google Scholar] [CrossRef]
  21. Clark, E.A.; Giltiay, N.V. CD22: A Regulator of Innate and Adaptive B Cell Responses and Autoimmunity. Front. Immunol. 2018, 9, 2235. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, K.; Wei, G.; Liu, D. CD19: A biomarker for B cell development, lymphoma diagnosis and therapy. Exp. Hematol. Oncol. 2012, 1, 36. [Google Scholar] [CrossRef] [Green Version]
  23. Sato, S.; Tuscano, J.M.; Inaoki, M.; Tedder, T.F. CD22 negatively and positively regulates signal transduction through the B lymphocyte antigen receptor. Semin. Immunol. 1998, 10, 287–297. [Google Scholar] [CrossRef]
  24. Tuscano, J.M.; Riva, A.; Toscano, S.N.; Tedder, T.F.; Kehrl, J.H. CD22 cross-linking generates B-cell antigen receptor-independent signals that activate the JNK/SAPK signaling cascade. Blood 1999, 94, 1382–1392. [Google Scholar] [CrossRef] [PubMed]
  25. Walker, J.A.; Smith, K.G. CD22: An inhibitory enigma. Immunology 2008, 123, 314–325. [Google Scholar] [CrossRef]
  26. Vaisitti, T.; Audrito, V.; Serra, S.; Buonincontri, R.; Sociali, G.; Mannino, E.; Pagnani, A.; Zucchetto, A.; Tissino, E.; Vitale, C.; et al. The enzymatic activities of CD38 enhance CLL growth and trafficking: Implications for therapeutic targeting. Leukemia 2015, 29, 356–368. [Google Scholar] [CrossRef]
  27. Sochacka-Ćwikła, A.; Mączyński, M.; Regiec, A. FDA-Approved Drugs for Hematological Malignancies—The Last Decade Review. Cancers 2021, 14, 87. [Google Scholar] [CrossRef] [PubMed]
  28. Puła, B.; Gołos, A.; Górniak, P.; Jamroziak, K. Overcoming Ibrutinib Resistance in Chronic Lymphocytic Leukemia. Cancers 2019, 11, 1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Marine, J.C.; Dawson, S.J.; Dawson, M.A. Non-genetic mechanisms of therapeutic resistance in cancer. Nat. Rev. Cancer 2020, 20, 743–756. [Google Scholar] [CrossRef] [PubMed]
  30. Roberts, A.W.; Wei, A.H.; Huang, D.C.S. BCL2 and MCL1 inhibitors for hematologic malignancies. Blood 2021, 138, 1120–1136. [Google Scholar] [CrossRef] [PubMed]
  31. Mohty, R.; Gauthier, J. Current combinatorial CAR T cell strategies with Bruton tyrosine kinase inhibitors and immune checkpoint inhibitors. Bone Marrow Transplant. 2021, 56, 2630–2636. [Google Scholar] [CrossRef] [PubMed]
  32. Schaefer, A.; Huang, Y.; Kittai, A.; Maakaron, J.E.; Saygin, C.; Brammer, J.; Penza, S.; Saad, A.; Jaglowski, S.M.; William, B.M. Cytopenias After CD19 Chimeric Antigen Receptor T-Cells (CAR-T) Therapy for Diffuse Large B-Cell Lymphomas or Transformed Follicular Lymphoma: A Single Institution Experience. Cancer Manag. Res. 2021, 13, 8901–8906. [Google Scholar] [CrossRef] [PubMed]
  33. Luo, C.; Wu, G.; Huang, X.; Ma, Y.; Zhang, Y.; Song, Q.; Xie, M.; Sun, Y.; Huang, Y.; Huang, Z.; et al. Efficacy and safety of new anti-CD20 monoclonal antibodies versus rituximab for induction therapy of CD20(+) B-cell non-Hodgkin lymphomas: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 3255. [Google Scholar] [CrossRef]
  34. Petrucci, M.T.; Vozella, F. The Anti-CD38 Antibody Therapy in Multiple Myeloma. Cells 2019, 8, 1629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Stam, R.W.; Den Boer, M.L.; Schneider, P.; de Boer, J.; Hagelstein, J.; Valsecchi, M.G.; de Lorenzo, P.; Sallan, S.E.; Brady, H.J.; Armstrong, S.A.; et al. Association of high-level MCL-1 expression with in vitro and in vivo prednisone resistance in MLL-rearranged infant acute lymphoblastic leukemia. Blood 2010, 115, 1018–1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rickert, R.C. New insights into pre-BCR and BCR signalling with relevance to B cell malignancies. Nat. Rev. Immunol. 2013, 13, 578–591. [Google Scholar] [CrossRef]
  37. Eswaran, J.; Sinclair, P.; Heidenreich, O.; Irving, J.; Russell, L.J.; Hall, A.; Calado, D.P.; Harrison, C.J.; Vormoor, J. The pre-B-cell receptor checkpoint in acute lymphoblastic leukaemia. Leukemia 2015, 29, 1623–1631. [Google Scholar] [CrossRef] [PubMed]
  38. Köhrer, S.; Havranek, O.; Seyfried, F.; Hurtz, C.; Coffey, G.P.; Kim, E.; Ten Hacken, E.; Jäger, U.; Vanura, K.; O’Brien, S.; et al. Pre-BCR signaling in precursor B-cell acute lymphoblastic leukemia regulates PI3K/AKT, FOXO1 and MYC, and can be targeted by SYK inhibition. Leukemia 2016, 30, 1246–1254. [Google Scholar] [CrossRef]
  39. Seyfried, F.; Stirnweiß, F.U.; Niedermayer, A.; Enzenmüller, S.; Hörl, R.L.; Münch, V.; Köhrer, S.; Debatin, K.M.; Meyer, L.H. Synergistic activity of combined inhibition of anti-apoptotic molecules in B-cell precursor ALL. Leukemia 2022, 36, 901–912. [Google Scholar] [CrossRef]
  40. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef]
  41. Vadillo, E.; Dorantes-Acosta, E.; Pelayo, R.; Schnoor, M. T cell acute lymphoblastic leukemia (T-ALL): New insights into the cellular origins and infiltration mechanisms common and unique among hematologic malignancies. Blood Rev. 2018, 32, 36–51. [Google Scholar] [CrossRef] [PubMed]
  42. Maitre, E.; Cornet, E.; Salaün, V.; Kerneves, P.; Chèze, S.; Repesse, Y.; Damaj, G.; Troussard, X. Immunophenotypic Analysis of Hairy Cell Leukemia (HCL) and Hairy Cell Leukemia-like (HCL-like) Disorders. Cancers 2022, 14, 1050. [Google Scholar] [CrossRef] [PubMed]
  43. Zaja, F.; Di Loreto, C.; Amoroso, V.; Salmaso, F.; Russo, D.; Silvestri, F.; Fanin, R.; Damiani, D.; Infanti, L.; Mariuzzi, L.; et al. BCL-2 immunohistochemical evaluation in B-cell chronic lymphocytic leukemia and hairy cell leukemia before treatment with fludarabine and 2-chloro-deoxy-adenosine. Leuk. Lymphoma 1998, 28, 567–572. [Google Scholar] [CrossRef] [PubMed]
  44. Batata, A.; Shen, B. Relationship between chronic lymphocytic leukemia and small lymphocytic lymphoma. A comparative study of membrane phenotypes in 270 cases. Cancer 1992, 70, 625–632. [Google Scholar] [CrossRef] [PubMed]
  45. Myklebust, J.H.; Brody, J.; Kohrt, H.E.; Kolstad, A.; Czerwinski, D.K.; Wälchli, S.; Green, M.R.; Trøen, G.; Liestøl, K.; Beiske, K.; et al. Distinct patterns of B-cell receptor signaling in non-Hodgkin lymphomas identified by single-cell profiling. Blood 2017, 129, 759–770. [Google Scholar] [CrossRef]
  46. Pangalis, G.A.; Angelopoulou, M.K.; Vassilakopoulos, T.P.; Siakantaris, M.P.; Kittas, C. B-chronic lymphocytic leukemia, small lymphocytic lymphoma, and lymphoplasmacytic lymphoma, including Waldenström’s macroglobulinemia: A clinical, morphologic, and biologic spectrum of similar disorders. Semin. Hematol. 1999, 36, 104–114. [Google Scholar]
  47. Zhang, S.; Chen, L.; Wang-Rodriguez, J.; Zhang, L.; Cui, B.; Frankel, W.; Wu, R.; Kipps, T.J. The onco-embryonic antigen ROR1 is expressed by a variety of human cancers. Am. J. Pathol. 2012, 181, 1903–1910. [Google Scholar] [CrossRef] [Green Version]
  48. Cho-Vega, J.H.; Rassidakis, G.Z.; Admirand, J.H.; Oyarzo, M.; Ramalingam, P.; Paraguya, A.; McDonnell, T.J.; Amin, H.M.; Medeiros, L.J. MCL-1 expression in B-cell non-Hodgkin’s lymphomas. Hum. Pathol. 2004, 35, 1095–1100. [Google Scholar] [CrossRef]
  49. Rajkumar, S.V.; Kumar, S. Multiple myeloma current treatment algorithms. Blood Cancer J. 2020, 10, 94. [Google Scholar] [CrossRef]
  50. Ilić, V.; Milosević-Jovcić, N.; Petrović, S.; Marković, D.; Bila, J.; Bosković, D.; Stefanović, G.; Marković, O.; Glibetić, M. Signaling status of IgG B cell receptor (IgG BCR) is indicative for an activated state of circulating B cells in multiple myeloma. Ann. Hematol. 2007, 86, 905–912. [Google Scholar] [CrossRef]
  51. Leite, L.A.; Kerbauy, D.M.; Kimura, E.; Yamamoto, M. Multiples aberrant phenotypes in multiple myeloma patient expressing CD56(-), CD28(+),CD19(+). Rev. Bras. Hematol. Hemoter. 2012, 34, 66–67. [Google Scholar] [CrossRef] [PubMed]
  52. Horna, P.; Nowakowski, G.; Endell, J.; Boxhammer, R. Comparative Assessment of Surface CD19 and CD20 Expression on B-Cell Lymphomas from Clinical Biopsies: Implications for Targeted Therapies. Blood 2019, 34, 627. [Google Scholar] [CrossRef]
  53. Radhakrishnan, V.S.; Lokireddy, P.; Parihar, M.; Prakash, P.S.; Menon, H. Mantle cell lymphoma: A clinical review of the changing treatment paradigms with the advent of novel therapies, and an insight into Indian data. Cancer Rep. 2021, 5, e1590. [Google Scholar] [CrossRef] [PubMed]
  54. Cheah, C.Y.; Zucca, E.; Rossi, D.; Habermann, T.M. Marginal zone lymphoma: Present status and future perspectives. Haematologica 2022, 107, 35–43. [Google Scholar] [CrossRef] [PubMed]
  55. Lai, R.; Arber, D.A.; Chang, K.L.; Wilson, C.S.; Weiss, L.M. Frequency of bcl-2 expression in non-Hodgkin’s lymphoma: A study of 778 cases with comparison of marginal zone lymphoma and monocytoid B-cell hyperplasia. Mod. Pathol. 1998, 11, 864–869. [Google Scholar] [PubMed]
  56. Wada, F.; Shimomura, Y.; Kamijo, K.; Yamashita, D.; Ohno, A.; Himeno, M.; Maruoka, H.; Hara, S.; Ishikawa, T. Prognostic impact of CD38 expression in relapsed or refractory diffuse large B-cell lymphoma and follicular lymphoma transformation. Leuk. Lymphoma 2022, 63, 1484–1487. [Google Scholar] [CrossRef]
  57. Diepstraten, S.T.; Chang, C.; Tai, L.; Gong, J.N.; Lan, P.; Dowell, A.C.; Taylor, G.S.; Strasser, A.; Kelly, G.L. BCL-W is dispensable for the sustained survival of select Burkitt lymphoma and diffuse large B-cell lymphoma cell lines. Blood Adv. 2020, 4, 356–366. [Google Scholar] [CrossRef] [Green Version]
  58. Nichols, G.L.; Stein, C.A. Modulation of the activity of Bcl-2 in Waldenstrom’s macroglobulinemia using antisense oligonucleotides. Semin. Oncol. 2003, 30, 297–299. [Google Scholar] [CrossRef]
  59. Pratt, G.; El-Sharkawi, D.; Kothari, J.; D’Sa, S.; Auer, R.; McCarthy, H.; Krishna, R.; Miles, O.; Kyriakou, C.; Owen, R. Diagnosis and management of Waldenström macroglobulinaemia-A British Society for Haematology guideline. Br. J. Haematol. 2022, 197, 171–187. [Google Scholar] [CrossRef]
  60. Lew, T.E.; Seymour, J.F. Clinical experiences with venetoclax and other pro-apoptotic agents in lymphoid malignancies: Lessons from monotherapy and chemotherapy combination. J. Hematol. Oncol. 2022, 15, 75. [Google Scholar] [CrossRef]
  61. Zhu, H.; Almasan, A. Development of venetoclax for therapy of lymphoid malignancies. Drug Des. Dev. Ther. 2017, 11, 685–694. [Google Scholar] [CrossRef] [PubMed]
  62. Hafezi, S.; Rahmani, M. Targeting BCL-2 in Cancer: Advances, Challenges, and Perspectives. Cancers 2021, 13, 1292. [Google Scholar] [CrossRef]
  63. Ferrarini, I.; Rigo, A.; Visco, C. The mitochondrial anti-apoptotic dependencies of hematologic malignancies: From disease biology to advances in precision medicine. Haematologica 2022, 107, 790–802. [Google Scholar] [CrossRef]
  64. Wierda, W.G.; Allan, J.N.; Siddiqi, T.; Kipps, T.J.; Opat, S.; Tedeschi, A.; Badoux, X.C.; Kuss, B.J.; Jackson, S.; Moreno, C.; et al. Ibrutinib Plus Venetoclax for First-Line Treatment of Chronic Lymphocytic Leukemia: Primary Analysis Results From the Minimal Residual Disease Cohort of the Randomized Phase II CAPTIVATE Study. J. Clin. Oncol. 2021, 39, 3853–3865. [Google Scholar] [CrossRef] [PubMed]
  65. Jain, N.; Keating, M.; Thompson, P.; Ferrajoli, A.; Burger, J.; Borthakur, G.; Takahashi, K.; Estrov, Z.; Fowler, N.; Kadia, T.; et al. Ibrutinib and Venetoclax for First-Line Treatment of CLL. N. Engl. J. Med. 2019, 380, 2095–2103. [Google Scholar] [CrossRef]
  66. Davids, M.S.; Roberts, A.W.; Seymour, J.F.; Pagel, J.M.; Kahl, B.S.; Wierda, W.G.; Puvvada, S.; Kipps, T.J.; Anderson, M.A.; Salem, A.H.; et al. Phase I First-in-Human Study of Venetoclax in Patients With Relapsed or Refractory Non-Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 826–833. [Google Scholar] [CrossRef] [Green Version]
  67. Wang, M.; Ramchandren, R.; Chen, R.; Karlin, L.; Chong, G.; Jurczak, W.; Wu, K.L.; Bishton, M.; Collins, G.P.; Eliadis, P.; et al. Concurrent ibrutinib plus venetoclax in relapsed/refractory mantle cell lymphoma: The safety run-in of the phase 3 SYMPATICO study. J. Hematol. Oncol. 2021, 14, 179. [Google Scholar] [CrossRef] [PubMed]
  68. Kumar, S.K.; Harrison, S.J.; Cavo, M.; de la Rubia, J.; Popat, R.; Gasparetto, C.; Hungria, V.; Salwender, H.; Suzuki, K.; Kim, I.; et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2020, 21, 1630–1642. [Google Scholar] [CrossRef] [PubMed]
  69. Morschhauser, F.; Feugier, P.; Flinn, I.W.; Gasiorowski, R.; Greil, R.; Illés, Á.; Johnson, N.A.; Larouche, J.F.; Lugtenburg, P.J.; Patti, C.; et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood 2021, 137, 600–609. [Google Scholar] [CrossRef]
  70. Del Gaizo Moore, V.; Schlis, K.D.; Sallan, S.E.; Armstrong, S.A.; Letai, A. BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia. Blood 2008, 111, 2300–2309. [Google Scholar] [CrossRef] [Green Version]
  71. Alford, S.E.; Kothari, A.; Loeff, F.C.; Eichhorn, J.M.; Sakurikar, N.; Goselink, H.M.; Saylors, R.L.; Jedema, I.; Falkenburg, J.H.; Chambers, T.C. BH3 Inhibitor Sensitivity and Bcl-2 Dependence in Primary Acute Lymphoblastic Leukemia Cells. Cancer Res. 2015, 75, 1366–1375. [Google Scholar] [CrossRef] [PubMed]
  72. Gibson, A.; Trabal, A.; McCall, D.; Khazal, S.; Toepfer, L.; Bell, D.H.; Roth, M.; Mahadeo, K.M.; Nunez, C.; Short, N.J.; et al. Venetoclax for Children and Adolescents with Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma. Cancers 2021, 14, 150. [Google Scholar] [CrossRef] [PubMed]
  73. Pullarkat, V.A.; Lacayo, N.J.; Jabbour, E.; Rubnitz, J.E.; Bajel, A.; Laetsch, T.W.; Leonard, J.; Colace, S.I.; Khaw, S.L.; Fleming, S.A.; et al. Venetoclax and Navitoclax in Combination with Chemotherapy in Patients with Relapsed or Refractory Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma. Cancer Discov. 2021, 11, 1440–1453. [Google Scholar] [CrossRef] [PubMed]
  74. Hunger, S.P.; Raetz, E.A. How I treat relapsed acute lymphoblastic leukemia in the pediatric population. Blood 2020, 136, 1803–1812. [Google Scholar] [CrossRef]
  75. Jain, N.; Stevenson, K.E.; Winer, E.S.; Garcia, J.S.; Stone, R.M.; Jabbour, E.; Ravandi, F.; Stewart, J.M.; Legg, D.R.; Kantarjian, H.M.; et al. A Multicenter Phase I Study Combining Venetoclax with Mini-Hyper-CVD in Older Adults with Untreated and Relapsed/Refractory Acute Lymphoblastic Leukemia. Blood 2019, 134, 3867. [Google Scholar] [CrossRef]
  76. Tam, C.S.; Verner, E.; Lasica, M.; Arbelaez, A.; Browett, P.J.; Soumerai, J.D.; Hilger, J.; Fang, Y.; Huang, J.; Simpson, D.; et al. Preliminary Safety and Efficacy Data from Patients (Pts) with Relapsed/Refractory (R/R) B-Cell Malignancies Treated with the Novel B-Cell Lymphoma 2 (BCL2) Inhibitor BGB-11417 in Monotherapy or in Combination with Zanubrutinib. Blood 2021, 138, 1419. [Google Scholar] [CrossRef]
  77. Sun, M.; Qi, J.; Chen, Z.; Zhang, H.; Song, Y.; Shen, A.; Liu, H.; Huang, J.; Zhou, F.; Jin, J.; et al. A Phase 1 Study to Evaluate the Safety, Pharmacokinetics (PK) and Pharmacodynamics (PD) of Lisaftoclax (APG-2575), a Novel BCL-2 Inhibitor (BCL-2i), in Patients (pts) with Certain Relapsed or Refractory (R/R) Hematologic Malignancies (HMs). Blood 2021, 138, 3730. [Google Scholar] [CrossRef]
  78. Yap, J.L.; Chen, L.; Lanning, M.E.; Fletcher, S. Expanding the Cancer Arsenal with Targeted Therapies: Disarmament of the Antiapoptotic Bcl-2 Proteins by Small Molecules. J. Med. Chem. 2017, 60, 821–838. [Google Scholar] [CrossRef]
  79. Wan, Y.; Dai, N.; Tang, Z.; Fang, H. Small-molecule Mcl-1 inhibitors: Emerging anti-tumor agents. Eur. J. Med. Chem. 2018, 146, 471–482. [Google Scholar] [CrossRef]
  80. Hird, A.W.; Tron, A.E. Recent advances in the development of Mcl-1 inhibitors for cancer therapy. Pharmacol. Ther. 2019, 198, 59–67. [Google Scholar] [CrossRef]
  81. Caenepeel, S.; Brown, S.P.; Belmontes, B.; Moody, G.; Keegan, K.S.; Chui, D.; Whittington, D.A.; Huang, X.; Poppe, L.; Cheng, A.C.; et al. AMG 176, a Selective MCL1 Inhibitor, Is Effective in Hematologic Cancer Models Alone and in Combination with Established Therapies. Cancer Discov. 2018, 8, 1582–1597. [Google Scholar] [CrossRef]
  82. Caenepeel, S.; Karen, R.; Belmontes, B.; Verlinsky, A.; Tan, H.; Yang, Y.; Chen, X.; Li, K.; Allen, J.; Wahlstrom, J.; et al. Discovery and preclinical evaluation of AMG 397, a potent, selective and orally bioavailable MCL1 inhibitor. Cancer Res. 2020, 180, 6218. [Google Scholar] [CrossRef]
  83. Szlavik, Z.; Csekei, M.; Paczal, A.; Szabo, Z.B.; Sipos, S.; Radics, G.; Proszenyak, A.; Balint, B.; Murray, J.; Davidson, J.; et al. Discovery of S64315, a Potent and Selective Mcl-1 Inhibitor. J. Med. Chem. 2020, 63, 13762–13795. [Google Scholar] [CrossRef] [PubMed]
  84. Tron, A.E.; Belmonte, M.A.; Adam, A.; Aquila, B.M.; Boise, L.H.; Chiarparin, E.; Cidado, J.; Embrey, K.J.; Gangl, E.; Gibbons, F.D.; et al. Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia. Nat. Commun. 2018, 9, 5341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Deak, D.; Pop, C.; Zimta, A.A.; Jurj, A.; Ghiaur, A.; Pasca, S.; Teodorescu, P.; Dascalescu, A.; Antohe, I.; Ionescu, B.; et al. Let’s Talk About BiTEs and Other Drugs in the Real-Life Setting for B-Cell Acute Lymphoblastic Leukemia. Front. Immunol. 2019, 10, 2856. [Google Scholar] [CrossRef] [PubMed]
  86. Hodby, K.A.; Marks, D.I. Recent Advances in the Management of Acute Lymphoblastic Leukaemia. Curr. Treat. Options Oncol. 2020, 21, 23. [Google Scholar] [CrossRef] [PubMed]
  87. Ratti, S.; Lonetti, A.; Follo, M.Y.; Paganelli, F.; Martelli, A.M.; Chiarini, F.; Evangelisti, C. B-ALL Complexity: Is Targeted Therapy Still A Valuable Approach for Pediatric Patients? Cancers 2020, 12, 3498. [Google Scholar] [CrossRef] [PubMed]
  88. Si Lim, S.J.; Grupp, S.A.; DiNofia, A.M. Tisagenlecleucel for treatment of children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia. Pediatr. Blood Cancer 2021, 68, e29123. [Google Scholar] [CrossRef] [PubMed]
  89. Zinzani, P.L.; Minotti, G. Anti-CD19 monoclonal antibodies for the treatment of relapsed or refractory B-cell malignancies: A narrative review with focus on diffuse large B-cell lymphoma. J. Cancer Res. Clin. Oncol. 2022, 148, 177–190. [Google Scholar] [CrossRef]
  90. Sanford, D.S.; Wierda, W.G.; Burger, J.A.; Keating, M.J.; O’Brien, S.M. Three newly approved drugs for chronic lymphocytic leukemia: Incorporating ibrutinib, idelalisib, and obinutuzumab into clinical practice. Clin. Lymphoma Myeloma Leuk. 2015, 15, 385–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Bewarder, M.; Stilgenbauer, S.; Thurner, L.; Kaddu-Mulindwa, D. Current Treatment Options in CLL. Cancers 2021, 13, 2468. [Google Scholar] [CrossRef] [PubMed]
  92. Eichhorst, B.; Robak, T.; Montserrat, E.; Ghia, P.; Niemann, C.U.; Kater, A.P.; Gregor, M.; Cymbalista, F.; Buske, C.; Hillmen, P.; et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2021, 32, 23–33. [Google Scholar] [CrossRef] [PubMed]
  93. Hallek, M.; Al-Sawaf, O. Chronic lymphocytic leukemia: 2022 update on diagnostic and therapeutic procedures. Am. J. Hematol. 2021, 96, 1679–1705. [Google Scholar] [CrossRef]
  94. Patel, K.; Pagel, J.M. Current and future treatment strategies in chronic lymphocytic leukemia. J. Hematol. Oncol. 2021, 14, 69. [Google Scholar] [CrossRef]
  95. Marchetti, M.; Rivela, P.; Bertassello, C.; Canicattì, M. Comparative Clinical Value of Pharmacologic Therapies for B-Cell Chronic Lymphocytic Leukemia: An Umbrella Analysis. J. Clin. Med. 2022, 11, 1868. [Google Scholar] [CrossRef] [PubMed]
  96. Kay, N.E.; Hampel, P.J.; Van Dyke, D.L.; Parikh, S.A. CLL update 2022: A continuing evolution in care. Blood Rev 2022, 54, 100930. [Google Scholar] [CrossRef] [PubMed]
  97. Small, S.; Ma, S. Frontline Treatment for Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL/SLL): Targeted Therapy vs. Chemoimmunotherapy. Curr. Hematol. Malig. Rep. 2021, 16, 325–335. [Google Scholar] [CrossRef] [PubMed]
  98. Sullivan-Chang, L.; O’Donnell, R.T.; Tuscano, J.M. Targeting CD22 in B-cell malignancies: Current status and clinical outlook. BioDrugs 2013, 27, 293–304. [Google Scholar] [CrossRef] [PubMed]
  99. Kreitman, R.J.; Dearden, C.; Zinzani, P.L.; Delgado, J.; Robak, T.; le Coutre, P.D.; Gjertsen, B.T.; Troussard, X.; Roboz, G.J.; Karlin, L.; et al. Moxetumomab pasudotox in heavily pre-treated patients with relapsed/refractory hairy cell leukemia (HCL): Long-term follow-up from the pivotal trial. J. Hematol. Oncol. 2021, 14, 35. [Google Scholar] [CrossRef] [PubMed]
  100. Dwivedi, S.; Rendón-Huerta, E.P.; Ortiz-Navarrete, V.; Montaño, L.F. CD38 and Regulation of the Immune Response Cells in Cancer. J. Oncol. 2021, 2021, 6630295. [Google Scholar] [CrossRef]
  101. Jiao, Y.; Yi, M.; Xu, L.; Chu, Q.; Yan, Y.; Luo, S.; Wu, K. CD38: Targeted therapy in multiple myeloma and therapeutic potential for solid cancers. Expert Opin. Investig. Drugs 2020, 29, 1295–1308. [Google Scholar] [CrossRef] [PubMed]
  102. Abramson, H.N. B-Cell Maturation Antigen (BCMA) as a Target for New Drug Development in Relapsed and/or Refractory Multiple Myeloma. Int. J. Mol. Sci. 2020, 21, 5192. [Google Scholar] [CrossRef] [PubMed]
  103. Hanel, W.; Epperla, N. Evolving therapeutic landscape in follicular lymphoma: A look at emerging and investigational therapies. J. Hematol. Oncol. 2021, 14, 104. [Google Scholar] [CrossRef]
  104. Pongas, G.; Cheson, B. Recent Advances in the Management of Patients with Relapsed/Refractory Follicular Lymphoma. Blood Lymphat. Cancer 2021, 11, 55–66. [Google Scholar] [CrossRef] [PubMed]
  105. Cahill, K.E.; Smith, S.M. Follicular Lymphoma: A Focus on Current and Emerging Therapies. Oncology 2022, 36, 97–106. [Google Scholar] [PubMed]
  106. Qualls, D.; Salles, G. Prospects in the management of patients with follicular lymphoma beyond first-line therapy. Haematologica 2022, 107, 19–34. [Google Scholar] [CrossRef]
  107. Narkhede, M.; Cheson, B.D. Copanlisib in the treatment of non-Hodgkin lymphoma. Future Oncol. 2020, 16, 1947–1955. [Google Scholar] [CrossRef]
  108. Salles, G.; Gopal, A.K.; Minnema, M.C.; Wakamiya, K.; Feng, H.; Schecter, J.M.; Wang, M. Phase 2 Study of Daratumumab in Relapsed/Refractory Mantle-Cell Lymphoma, Diffuse Large B-Cell Lymphoma, and Follicular Lymphoma. Clin. Lymphoma Myeloma Leuk. 2019, 19, 275–284. [Google Scholar] [CrossRef]
  109. Pu, J.J.; Savani, M.; Huang, N.; Epner, E.M. Mantle cell lymphoma management trends and novel agents: Where are we going? Ther. Adv. Hematol. 2022, 13, 20406207221080743. [Google Scholar] [CrossRef] [PubMed]
  110. Alderuccio, J.P.; Kahl, B.S. Current Treatments in Marginal Zone Lymphoma. Oncology 2022, 36, 206–215. [Google Scholar]
  111. Chilakamarri, N.O.K.; Brem, E.A. Current and Future Therapies for Marginal Zone Lymphoma. Oncol. Haematol. 2022, 2022, 18. [Google Scholar] [CrossRef]
  112. Opat, S.; Tedeschi, A.; Linton, K.; McKay, P.; Hu, B.; Chan, H.; Jin, J.; Sobieraj-Teague, M.; Zinzani, P.L.; Coleman, M.; et al. The MAGNOLIA Trial: Zanubrutinib, a Next-Generation Bruton Tyrosine Kinase Inhibitor, Demonstrates Safety and Efficacy in Relapsed/Refractory Marginal Zone Lymphoma. Clin. Cancer Res. 2021, 27, 6323–6332. [Google Scholar] [CrossRef] [PubMed]
  113. Micallef, I.N.; Maurer, M.J.; Wiseman, G.A.; Nikcevich, D.A.; Kurtin, P.J.; Cannon, M.W.; Perez, D.G.; Soori, G.S.; Link, B.K.; Habermann, T.M.; et al. Epratuzumab with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy in patients with previously untreated diffuse large B-cell lymphoma. Blood 2011, 118, 4053–4061. [Google Scholar] [CrossRef] [Green Version]
  114. Cheson, B.D.; Nowakowski, G.; Salles, G. Diffuse large B-cell lymphoma: New targets and novel therapies. Blood Cancer J. 2021, 11, 68. [Google Scholar] [CrossRef] [PubMed]
  115. Spinner, M.A.; Advani, R.H. Current Frontline Treatment of Diffuse Large B-Cell Lymphoma. Oncology 2022, 36, 51–58. [Google Scholar]
  116. Davis, J.A.; Shockley, A.; Glode, A.E. Newly approved anti-CD19 monoclonal antibodies for the treatment of relapsed or refractory diffuse large B-cell lymphoma. J. Oncol. Pharm. Pract. 2022, 28, 686–690. [Google Scholar] [CrossRef]
  117. Kahl, B.S.; Hamadani, M.; Radford, J.; Carlo-Stella, C.; Caimi, P.; Reid, E.; Feingold, J.M.; Ardeshna, K.M.; Solh, M.; Heffner, L.T.; et al. A Phase I Study of ADCT-402 (Loncastuximab Tesirine), a Novel Pyrrolobenzodiazepine-Based Antibody-Drug Conjugate, in Relapsed/Refractory B-Cell Non-Hodgkin Lymphoma. Clin. Cancer Res. 2019, 25, 6986–6994. [Google Scholar] [CrossRef] [Green Version]
  118. Castillo, J.J.; Advani, R.H.; Branagan, A.R.; Buske, C.; Dimopoulos, M.A.; D’Sa, S.; Kersten, M.J.; Leblond, V.; Minnema, M.C.; Owen, R.G.; et al. Consensus treatment recommendations from the tenth International Workshop for Waldenström Macroglobulinaemia. Lancet Haematol. 2020, 7, e827–e837. [Google Scholar] [CrossRef]
  119. Grimont, C.N.; Castillo Almeida, N.E.; Gertz, M.A. Current and Emerging Treatments for Waldenström Macroglobulinemia. Acta Haematol. 2021, 144, 146–157. [Google Scholar] [CrossRef] [PubMed]
  120. Thomas, S.K. SOHO State of the Art Updates and Next Questions: Waldenström Macroglobulinemia-2021 Update on Management and Future Directions. Clin. Lymphoma Myeloma Leuk. 2022, 22, 347–355. [Google Scholar] [CrossRef]
  121. Gertz, M.A. Waldenström macroglobulinemia: 2021 update on diagnosis, risk stratification, and management. Am. J. Hematol. 2021, 96, 258–269. [Google Scholar] [CrossRef]
  122. Castillo, J.J.; Treon, S.P. Management of Waldenström macroglobulinemia in 2020. Hematol. Am. Soc. Hematol. Educ. Program 2020, 1, 372–379. [Google Scholar] [CrossRef] [PubMed]
  123. Li, J.; Zhou, K.; Nian, W.; Cui, G.; Wang, J.; Zhang, X.; Cen, H.; Li, F.; Yi, S.; Feng, R.; et al. A phase Ib/II study of lisaftoclax (APG-2575), a novel BCL-2 inhibitor (BCL-2i), in patients (pts) with relapsed/refractory chronic lymphocytic leukemia or small lymphocytic lymphoma (R/R CLL/SLL). J. Clin. Oncol. 2022, 40, 7543. [Google Scholar] [CrossRef]
  124. Bhagwat, N.; Grego, A.; Gowen-MacDonald, W.; Wang, M.; Cowart, M.; Wu, X.; Zhuo, J.; Combs, A.; Ruggeri, B.; Scherle, P.; et al. Preclinical characterization of PRT1419, a potent, selective and orally available inhibitor of MCL1. Cancer Res. 2021, 81, 983. [Google Scholar] [CrossRef]
  125. Dhillon, S. Orelabrutinib: First Approval. Drugs 2021, 81, 503–507. [Google Scholar] [CrossRef] [PubMed]
  126. Jurczak, W.; Zinzani, P.L.L.; Cunningham, D.; Azoulay, M.; Huang, W.; Xu, W.; Ribrag, V. Coastal: A phase 3 study of the PI3Kδ inhibitor zandelisib with rituximab (R) versus immunochemotherapy in patients with relapsed indolent non-Hodgkin’s lymphoma (iNHL). J. Clin. Oncol. 2021, 39, 7573. [Google Scholar] [CrossRef]
  127. Davids, M.S.; Kim, H.T.; Nicotra, A.; Savell, A.; Francoeur, K.; Hellman, J.M.; Bazemore, J.; Miskin, H.P.; Sportelli, P.; Stampleman, L.; et al. Umbralisib in combination with ibrutinib in patients with relapsed or refractory chronic lymphocytic leukaemia or mantle cell lymphoma: A multicentre phase 1-1b study. Lancet Haematol. 2019, 6, e38–e47. [Google Scholar] [CrossRef]
  128. Budde, L.E.; Assouline, S.; Sehn, L.H.; Schuster, S.J.; Yoon, S.S.; Yoon, D.H.; Matasar, M.J.; Bosch, F.; Kim, W.S.; Nastoupil, L.J.; et al. Single-Agent Mosunetuzumab Shows Durable Complete Responses in Patients With Relapsed or Refractory B-Cell Lymphomas: Phase I Dose-Escalation Study. J. Clin. Oncol. 2022, 40, 481–491. [Google Scholar] [CrossRef]
  129. Bannerji, R.; Allan, J.N.; Arnason, J.E.; Brown, J.R.; Advani, R.H.; Barnes, J.A.; Ansell, S.M.; O’Brien, S.M.; Chavez, J.; Duell, J.; et al. Clinical Activity of REGN1979, a Bispecific Human, Anti-CD20 x Anti-CD3 Antibody, in Patients with Relapsed/Refractory (R/R) B-Cell Non-Hodgkin Lymphoma (B-NHL). Blood 2019, 134, 762. [Google Scholar] [CrossRef]
  130. Van der Horst, H.J.; de Jonge, A.V.; Hiemstra, I.H.; Gelderloos, A.T.; Berry, D.; Hijmering, N.J.; van Essen, H.F.; de Jong, D.; Chamuleau, M.E.D.; Zweegman, S.; et al. Epcoritamab induces potent anti-tumor activity against malignant B-cells from patients with DLBCL, FL and MCL, irrespective of prior CD20 monoclonal antibody treatment. Blood Cancer J. 2021, 11, 38. [Google Scholar] [CrossRef] [PubMed]
  131. Engelberts, P.J.; Hiemstra, I.H.; de Jong, B.; Schuurhuis, D.H.; Meesters, J.; Beltran Hernandez, I.; Oostindie, S.C.; Neijssen, J.; van den Brink, E.N.; Horbach, G.J.; et al. DuoBody-CD3xCD20 induces potent T-cell-mediated killing of malignant B cells in preclinical models and provides opportunities for subcutaneous dosing. eBioMedicine 2020, 52, 102625. [Google Scholar] [CrossRef] [PubMed]
  132. Hutchings, M.; Morschhauser, F.; Iacoboni, G.; Carlo-Stella, C.; Offner, F.C.; Sureda, A.; Salles, G.; Martínez-Lopez, J.; Crump, M.; Thomas, D.N.; et al. Glofitamab, a Novel, Bivalent CD20-Targeting T-Cell-Engaging Bispecific Antibody, Induces Durable Complete Remissions in Relapsed or Refractory B-Cell Lymphoma: A Phase I Trial. J. Clin. Oncol. 2021, 39, 1959–1970. [Google Scholar] [CrossRef] [PubMed]
  133. Salles, G.; Duell, J.; González Barca, E.; Tournilhac, O.; Jurczak, W.; Liberati, A.M.; Nagy, Z.; Obr, A.; Gaidano, G.; André, M.; et al. Tafasitamab plus lenalidomide in relapsed or refractory diffuse large B-cell lymphoma (L-MIND): A multicentre, prospective, single-arm, phase 2 study. Lancet Oncol. 2020, 21, 978–988. [Google Scholar] [CrossRef] [PubMed]
  134. Shah, N.N.; Johnson, B.D.; Schneider, D.; Zhu, F.; Szabo, A.; Keever-Taylor, C.A.; Krueger, W.; Worden, A.A.; Kadan, M.J.; Yim, S.; et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: A phase 1 dose escalation and expansion trial. Nat. Med. 2020, 26, 1569–1575. [Google Scholar] [CrossRef]
  135. Liu, E.; Tong, Y.; Dotti, G.; Shaim, H.; Savoldo, B.; Mukherjee, M.; Orange, J.; Wan, X.; Lu, X.; Reynolds, A.; et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2018, 32, 520–531. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef] [PubMed]
  137. Dickinson, M.; Hamad, N.; Bryant, C.E.; Borthakur, G.; Hosing, C.; Shook, D.; Tan, J.; Rajangam, K.; Liu, H.; Kennedy, G.A.; et al. A Phase 1 Study of NKX019, a CD19 Chimeric Antigen Receptor Natural Killer (CAR NK) Cell Therapy, in Subjects with B-Cell Malignancies. Blood 2021, 138, 3868. [Google Scholar] [CrossRef]
  138. Cheng, Q.; Tan, J.; Liu, R.; Kang, L.; Zhang, Y.; Wang, E.; Li, Y.; Zhang, J.; Xiao, H.; Xu, N.; et al. CD20-specific chimeric antigen receptor-expressing T cells as salvage therapy in rituximab-refractory/relapsed B-cell non-Hodgkin lymphoma. Cytotherapy 2022, 24, 1026–1034. [Google Scholar] [CrossRef] [PubMed]
  139. Fry, T.J.; Shah, N.N.; Orentas, R.J.; Stetler-Stevenson, M.; Yuan, C.M.; Ramakrishna, S.; Wolters, P.; Martin, S.; Delbrook, C.; Yates, B.; et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 2018, 24, 20–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Shah, N.N.; Zhu, F.; Taylor, C.; Schneider, D.; Krueger, W.; Worden, A.; Yim, S.; Fenske, T.S.; Hamadani, M.; Johnson, B.; et al. A Phase 1 Study with Point-of-Care Manufacturing of Dual Targeted, Tandem Anti-CD19, Anti-CD20 Chimeric Antigen Receptor Modified T (CAR-T) Cells for Relapsed, Refractory, Non-Hodgkin Lymphoma. Blood 2018, 132, 4193. [Google Scholar] [CrossRef]
  141. Osborne, W.; Marzolini, M.; Tholouli, E.; Ramakrishnan, A.; Bachier, C.R.; McSweeney, P.A.; Irvine, D.; Zhang, M.; Al-Hajj, M.A.; Pule, M.; et al. Phase I Alexander study of AUTO3, the first CD19/22 dual targeting CAR T cell therapy, with pembrolizumab in patients with relapsed/refractory (r/r) DLBCL. J. Clin. Oncol. 2020, 38, 8001. [Google Scholar] [CrossRef]
  142. Spiegel, J.Y.; Patel, S.; Muffly, L.; Hossain, N.M.; Oak, J.; Baird, J.H.; Frank, M.J.; Shiraz, P.; Sahaf, B.; Craig, J.; et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: A phase 1 trial. Nat. Med. 2021, 27, 1419–1431. [Google Scholar] [CrossRef]
  143. Shalabi, H.; Nellan, A.; Shah, N.N.; Gust, J. Immunotherapy Associated Neurotoxicity in Pediatric Oncology. Front. Oncol. 2022, 12, 836452. [Google Scholar] [CrossRef] [PubMed]
  144. Vogl, D.T.; Kaufman, J.L.; Holstein, S.A.; Atrash, S.; Nadeem, O.; Janakiram, M.; Suryanarayan, K.; Liu, Y.; Collins, S.; Parot, X.; et al. Modakafusp Alfa (TAK-573), an Immunocytokine, Shows Clinical Activity in Patients with Relapsed/Refractory Multiple Myeloma; Updated Results from a First-in-Human Phase 1 Study. Blood 2021, 138, 898. [Google Scholar] [CrossRef]
  145. Kumar, S.A.M.; Dabovic, K.; Wang, J.; Anand, B.; Yuet, A.; Dholaria, B.; Roy, V. 447 Interim results of a phase 1 study of the novel engineered toxin body TAK-169 in patients with relapsed or refractory multiple myeloma. J. Immunother. Cancer 2021, 9, A475. [Google Scholar] [CrossRef]
  146. Byrd, J.C.; Hillmen, P.; Ghia, P.; Kater, A.P.; Chanan-Khan, A.; Furman, R.R.; O’Brien, S.; Yenerel, M.N.; Illés, A.; Kay, N.; et al. Acalabrutinib Versus Ibrutinib in Previously Treated Chronic Lymphocytic Leukemia: Results of the First Randomized Phase III Trial. J. Clin. Oncol. 2021, 39, 3441–3452. [Google Scholar] [CrossRef]
  147. Danilov, A.V.; Persky, D.O. Incorporating acalabrutinib, a selective next-generation Bruton tyrosine kinase inhibitor, into clinical practice for the treatment of haematological malignancies. Br. J. Haematol. 2021, 193, 15–25. [Google Scholar] [CrossRef]
  148. Zhu, S.; Jung, J.; Victor, E.; Arceo, J.; Gokhale, S.; Xie, P. Clinical Trials of the BTK Inhibitors Ibrutinib and Acalabrutinib in Human Diseases Beyond B Cell Malignancies. Front. Oncol. 2021, 11, 737943. [Google Scholar] [CrossRef]
  149. Ran, F.; Liu, Y.; Wang, C.; Xu, Z.; Zhang, Y.; Liu, Y.; Zhao, G.; Ling, Y. Review of the development of BTK inhibitors in overcoming the clinical limitations of ibrutinib. Eur. J. Med. Chem. 2022, 229, 114009. [Google Scholar] [CrossRef]
  150. Muñoz, J.; Wang, Y.; Jain, P.; Wang, M. Zanubrutinib in lymphoproliferative disorders: A comprehensive review. Ther. Adv. Hematol. 2022, 13, 20406207221093980. [Google Scholar] [CrossRef]
  151. Campbell, R.; Chong, G.; Hawkes, E.A. Novel Indications for Bruton’s Tyrosine Kinase Inhibitors, beyond Hematological Malignancies. J. Clin. Med. 2018, 7, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Von Suskil, M.; Sultana, K.N.; Elbezanti, W.O.; Al-Odat, O.S.; Chitren, R.; Tiwari, A.K.; Challagundla, K.B.; Srivastava, S.K.; Jonnalagadda, S.C.; Budak-Alpdogan, T.; et al. Bruton’s Tyrosine Kinase Targeting in Multiple Myeloma. Int. J. Mol. Sci. 2021, 22, 5707. [Google Scholar] [CrossRef]
  153. Wilson, W.H.; Wright, G.W.; Huang, D.W.; Hodkinson, B.; Balasubramanian, S.; Fan, Y.; Vermeulen, J.; Shreeve, M.; Staudt, L.M. Effect of ibrutinib with R-CHOP chemotherapy in genetic subtypes of DLBCL. Cancer Cell 2021, 39, 1643–1653.e3. [Google Scholar] [CrossRef] [PubMed]
  154. Gopal, A.K.; Schuster, S.J.; Fowler, N.H.; Trotman, J.; Hess, G.; Hou, J.Z.; Yacoub, A.; Lill, M.; Martin, P.; Vitolo, U.; et al. Ibrutinib as Treatment for Patients With Relapsed/Refractory Follicular Lymphoma: Results From the Open-Label, Multicenter, Phase II DAWN Study. J. Clin. Oncol. 2018, 36, 2405–2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Mozas, P.; Delgado, J. Zanubrutinib joins the CLL treatment buffet. Lancet Oncol. 2022, 23, 965–967. [Google Scholar] [CrossRef]
  156. Zinzani, P.L.; Mayer, J.; Auer, R.; Bijou, F.; de Oliveira, A.C.; Flowers, C.; Merli, M.; Bouabdallah, K.; Ganly, P.S.; Johnson, R.; et al. Zanubrutinib plus obinutuzumab (ZO) versus obinutuzumab (O) monotherapy in patients (pts) with relapsed or refractory (R/R) follicular lymphoma (FL): Primary analysis of the phase 2 randomized ROSEWOOD trial. J. Clin. Oncol. 2022, 40, 7510. [Google Scholar] [CrossRef]
  157. Yang, H.; Xiang, B.; Song, Y.; Zhang, H.; Zhao, W.; Zou, D.; Lv, F.; Guo, W.; Liu, A.; Li, C.; et al. Zanubrutinib monotherapy for relapsed or refractory non-germinal center diffuse large B-cell lymphoma. Blood Adv. 2022, 6, 1629–1636. [Google Scholar] [CrossRef]
  158. Ferrer, G.; Montserrat, E. Critical molecular pathways in CLL therapy. Mol. Med. 2018, 24, 9. [Google Scholar] [CrossRef]
  159. Byrd, J.C.; Smith, S.; Wagner-Johnston, N.; Sharman, J.; Chen, A.I.; Advani, R.; Augustson, B.; Marlton, P.; Renee Commerford, S.; Okrah, K.; et al. First-in-human phase 1 study of the BTK inhibitor GDC-0853 in relapsed or refractory B-cell NHL and CLL. Oncotarget 2018, 9, 13023–13035. [Google Scholar] [CrossRef] [Green Version]
  160. Phillips, T.J.; Michot, J.M.; Ribrag, V. Can Next-Generation PI3K Inhibitors Unlock the Full Potential of the Class in Patients With B-Cell Lymphoma? Clin. Lymphoma Myeloma Leuk. 2021, 21, 8–20.e3. [Google Scholar] [CrossRef] [PubMed]
  161. Hus, I.; Puła, B.; Robak, T. PI3K Inhibitors for the Treatment of Chronic Lymphocytic Leukemia: Current Status and Future Perspectives. Cancers 2022, 14, 1571. [Google Scholar] [CrossRef] [PubMed]
  162. Shah, A.; Barrientos, J.C. Oral PI3K-δ,γ Inhibitor for the Management of People with Chronic Lymphocytic Leukemia and Small Lymphocytic Lymphoma: A Narrative Review on Duvelisib. Onco Targets Ther. 2021, 14, 2109–2119. [Google Scholar] [CrossRef] [PubMed]
  163. Shouse, G.; Danilova, O.V.; Danilov, A.V. Current status of phosphoinotiside-3 kinase inhibitors in blood cancers. Curr. Opin. Oncol. 2022, 34, 540–545. [Google Scholar] [CrossRef] [PubMed]
  164. Le, T.; Jerel, D.; Bryan, L.J. Update on the role of copanlisib in hematologic malignancies. Ther. Adv. Hematol. 2021, 12, 20406207211006027. [Google Scholar] [CrossRef] [PubMed]
  165. Gopal, A.K.; Kahl, B.S.; de Vos, S.; Wagner-Johnston, N.D.; Schuster, S.J.; Jurczak, W.J.; Flinn, I.W.; Flowers, C.R.; Martin, P.; Viardot, A.; et al. PI3Kδ inhibition by idelalisib in patients with relapsed indolent lymphoma. N. Engl. J. Med. 2014, 370, 1008–1018. [Google Scholar] [CrossRef] [Green Version]
  166. Chauhan, A.F.; Cheson, B.D. Copanlisib in the Treatment of Relapsed Follicular Lymphoma: Utility and Experience from the Clinic. Cancer Manag. Res. 2021, 13, 677–692. [Google Scholar] [CrossRef]
  167. Casadei, B.; Argnani, L.; Broccoli, A.; Patti, C.; Stefani, P.M.; Cuneo, A.; Margiotta Casaluci, G.; Visco, C.; Gini, G.; Pane, F.; et al. Treatment with Idelalisib in Patients with Relapsed or Refractory Follicular Lymphoma: The Observational Italian Multicenter FolIdela Study. Cancers 2022, 14, 654. [Google Scholar] [CrossRef]
  168. Rodrigues, D.A.; Sagrillo, F.S.; Fraga, C.A.M. Duvelisib: A 2018 Novel FDA-Approved Small Molecule Inhibiting Phosphoinositide 3-Kinases. Pharmaceuticals 2019, 12, 69. [Google Scholar] [CrossRef] [Green Version]
  169. Tucker, N. Duvelisib for R/R Follicular Lymphoma Voluntarily Pulled from the US Market. Target. Oncol. 2021. Available online: https://www.targetedonc.com/view/duvelisib-for-r-r-follicular-lymphoma-voluntarily-pulled-from-the-us-market (accessed on 6 December 2021).
  170. Matasar, M.J.; Capra, M.; Özcan, M.; Lv, F.; Li, W.; Yañez, E.; Sapunarova, K.; Lin, T.; Jin, J.; Jurczak, W.; et al. Copanlisib plus rituximab versus placebo plus rituximab in patients with relapsed indolent non-Hodgkin lymphoma (CHRONOS-3): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2021, 22, 678–689. [Google Scholar] [CrossRef]
  171. Forero-Torres, A.; Ramchandren, R.; Yacoub, A.; Wertheim, M.S.; Edenfield, W.J.; Caimi, P.; Gutierrez, M.; Akard, L.; Escobar, C.; Call, J.; et al. Parsaclisib, a potent and highly selective PI3Kδ inhibitor, in patients with relapsed or refractory B-cell malignancies. Blood 2019, 133, 1742–1752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Coleman, M.; Belada, D.; Casasnovas, R.O.; Gressin, R.; Lee, H.P.; Mehta, A.; Munoz, J.; Verhoef, G.; Corrado, C.; DeMarini, D.J.; et al. Phase 2 study of parsaclisib (INCB050465), a highly selective, next-generation PI3Kδ inhibitor, in relapsed or refractory diffuse large B-cell lymphoma (CITADEL-202). Leuk Lymphoma 2021, 62, 368–376. [Google Scholar] [CrossRef] [PubMed]
  173. Joszt, L. Incyte Withdraws FDA Submission for Parsaclisib for FL, MZL, MCL. AJMC 2022. Available online: https://www.ajmc.com/view/incyte-withdraws-fda-submission-for-parsaclisib-for-fl-mzl-mcl (accessed on 28 January 2022).
  174. Pagel, J.M.; Soumerai, J.D.; Reddy, N.; Jagadeesh, D.; Stathis, A.; Asch, A.; Salman, H.; Kenkre, V.P.; Iasonos, A.; Llorin-Sangalang, J.; et al. Zandelisib with continuous or intermittent dosing as monotherapy or in combination with rituximab in patients with relapsed or refractory B-cell malignancy: A multicentre, first-in-patient, dose-escalation and dose-expansion, phase 1b trial. Lancet Oncol. 2022, 23, 1021–1030. [Google Scholar] [CrossRef] [PubMed]
  175. Maharaj, K.; Powers, J.J.; Achille, A.; Mediavilla-Varela, M.; Gamal, W.; Burger, K.L.; Fonseca, R.; Jiang, K.; Miskin, H.P.; Maryanski, D.; et al. The dual PI3Kδ/CK1ε inhibitor umbralisib exhibits unique immunomodulatory effects on CLL T cells. Blood Adv. 2020, 4, 3072–3084. [Google Scholar] [CrossRef]
  176. Burris, H.A., 3rd; Flinn, I.W.; Patel, M.R.; Fenske, T.S.; Deng, C.; Brander, D.M.; Gutierrez, M.; Essell, J.H.; Kuhn, J.G.; Miskin, H.P.; et al. Umbralisib, a novel PI3Kδ and casein kinase-1ε inhibitor, in relapsed or refractory chronic lymphocytic leukaemia and lymphoma: An open-label, phase 1, dose-escalation, first-in-human study. Lancet Oncol. 2018, 19, 486–496. [Google Scholar] [CrossRef] [PubMed]
  177. Mato, A.R.; Ghosh, N.; Schuster, S.J.; Lamanna, N.; Pagel, J.M.; Flinn, I.W.; Barrientos, J.C.; Rai, K.R.; Reeves, J.A.; Cheson, B.D.; et al. Phase 2 study of the safety and efficacy of umbralisib in patients with CLL who are intolerant to BTK or PI3Kδ inhibitor therapy. Blood 2021, 137, 2817–2826. [Google Scholar] [CrossRef]
  178. Lunning, M.; Vose, J.; Nastoupil, L.; Fowler, N.; Burger, J.A.; Wierda, W.G.; Schreeder, M.T.; Siddiqi, T.; Flowers, C.R.; Cohen, J.B.; et al. Ublituximab and umbralisib in relapsed/refractory B-cell non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood 2019, 134, 1811–1820. [Google Scholar] [CrossRef]
  179. Virgil, H. FDA Places Partial Clinical Hold on Studies Featuring Umbralisib and Ublituximab in CLL and Non-Hodgkin Lymphoma. Cancer Netwok 2022. Available online: https://www.cancerneetwork.com/view/fda-places-partial-clinical-hold-on-studies-featuring-umbralisib-and-ublituximab-in-cll-and-non-hodgkin-lymphoma (accessed on 1 February 2022).
  180. Lu, R.M.; Hwang, Y.C.; Liu, I.J.; Lee, C.C.; Tsai, H.Z.; Li, H.J.; Wu, H.C. Development of therapeutic antibodies for the treatment of diseases. J. Biomed. Sci. 2020, 27, 1. [Google Scholar] [CrossRef]
  181. Fishelson, Z.; Kirschfink, M. Complement C5b-9 and Cancer: Mechanisms of Cell Damage, Cancer Counteractions, and Approaches for Intervention. Front. Immunol. 2019, 10, 752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Chen, Z.; Kankala, R.K.; Yang, Z.; Li, W.; Xie, S.; Li, H.; Chen, A.Z.; Zou, L. Antibody-based drug delivery systems for cancer therapy: Mechanisms, challenges, and prospects. Theranostics 2022, 12, 3719–3746. [Google Scholar] [CrossRef]
  183. Hedrich, W.D.; Fandy, T.E.; Ashour, H.M.; Wang, H.; Hassan, H.E. Antibody-Drug Conjugates: Pharmacokinetic/Pharmacodynamic Modeling, Preclinical Characterization, Clinical Studies, and Lessons Learned. Clin. Pharmacokinet. 2018, 57, 687–703. [Google Scholar] [CrossRef] [PubMed]
  184. Lejeune, M.; Köse, M.C.; Duray, E.; Einsele, H.; Beguin, Y.; Caers, J. Bispecific, T-Cell-Recruiting Antibodies in B-Cell Malignancies. Front. Immunol. 2020, 11, 762. [Google Scholar] [CrossRef] [PubMed]
  185. Morcos, P.N.; Li, J.; Hosseini, I.; Li, C.C. Quantitative Clinical Pharmacology of T-Cell Engaging Bispecifics: Current Perspectives and Opportunities. Clin. Transl. Sci. 2021, 14, 75–85. [Google Scholar] [CrossRef]
  186. Beck, A.; Goetsch, L.; Dumontet, C.; Corvaïa, N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat. Rev. Drug Discov. 2017, 16, 315–337. [Google Scholar] [CrossRef]
  187. Spiess, C.; Zhai, Q.; Carter, P.J. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol. Immunol. 2015, 67, 95–106. [Google Scholar] [CrossRef]
  188. Keane, J.T.; Posey, A.D., Jr. Chimeric Antigen Receptors Expand the Repertoire of Antigenic Macromolecules for Cellular Immunity. Cells 2021, 10, 3356. [Google Scholar] [CrossRef] [PubMed]
  189. Abrantes, R.; Duarte, H.O.; Gomes, C.; Wälchli, S.; Reis, C.A. CAR-Ts: New perspectives in cancer therapy. FEBS Lett. 2022, 596, 403–416. [Google Scholar] [CrossRef] [PubMed]
  190. Fischer, J.W.; Bhattarai, N. CAR-T Cell Therapy: Mechanism, Management, and Mitigation of Inflammatory Toxicities. Front. Immunol. 2021, 12, 693016. [Google Scholar] [CrossRef] [PubMed]
  191. Moreno, C.; Haynie, C.; Johnson, A.; Weber, K.S. Alternative CAR Therapies: Recent Approaches in Engineering Chimeric Antigen Receptor Immune Cells to Combat Cancer. Biomedicines 2022, 10, 1493. [Google Scholar] [CrossRef] [PubMed]
  192. Lim, S.H.; Beers, S.A.; French, R.R.; Johnson, P.W.; Glennie, M.J.; Cragg, M.S. Anti-CD20 monoclonal antibodies: Historical and future perspectives. Haematologica 2010, 95, 135–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Tuscano, J.M.; Maverakis, E.; Groshen, S.; Tsao-Wei, D.; Luxardi, G.; Merleev, A.A.; Beaven, A.; DiPersio, J.F.; Popplewell, L.; Chen, R.; et al. A Phase I Study of the Combination of Rituximab and Ipilimumab in Patients with Relapsed/Refractory B-Cell Lymphoma. Clin. Cancer Res. 2019, 25, 7004–7013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Marshall, M.J.E.; Stopforth, R.J.; Cragg, M.S. Therapeutic Antibodies: What Have We Learnt from Targeting CD20 and Where Are We Going? Front. Immunol. 2017, 8, 1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Singh, S.; Kumar, N.K.; Dwiwedi, P.; Charan, J.; Kaur, R.; Sidhu, P.; Chugh, V.K. Monoclonal Antibodies: A Review. Curr. Clin. Pharmacol. 2018, 13, 85–99. [Google Scholar] [CrossRef]
  196. Bannerji, R.; Arnason, J.E.; Advani, R.H.; Brown, J.R.; Allan, J.N.; Ansell, S.M.; Barnes, J.A.; O’Brien, S.M.; Chávez, J.C.; Duell, J.; et al. Odronextamab, a human CD20×CD3 bispecific antibody in patients with CD20-positive B-cell malignancies (ELM-1): Results from the relapsed or refractory non-Hodgkin lymphoma cohort in a single-arm, multicentre, phase 1 trial. Lancet Haematol. 2022, 9, e327–e339. [Google Scholar] [CrossRef]
  197. Zhao, Z.; Chen, Y.; Francisco, N.M.; Zhang, Y.; Wu, M. The application of CAR-T cell therapy in hematological malignancies: Advantages and challenges. Acta Pharm. Sin. B 2018, 8, 539–551. [Google Scholar] [CrossRef]
  198. Aleksandrova, K.; Leise, J.; Priesner, C.; Melk, A.; Kubaink, F.; Abken, H.; Hombach, A.; Aktas, M.; Essl, M.; Bürger, I.; et al. Functionality and Cell Senescence of CD4/CD8-Selected CD20 CAR T Cells Manufactured Using the Automated CliniMACS Prodigy® Platform. Transfus. Med. Hemother. 2019, 46, 47–54. [Google Scholar] [CrossRef]
  199. Naddafi, F.; Davami, F. Anti-CD19 Monoclonal Antibodies: A New Approach to Lymphoma Therapy. Int. J. Mol. Cell. Med. 2015, 4, 143–151. [Google Scholar]
  200. Watkins, M.P.; Bartlett, N.L. CD19-targeted immunotherapies for treatment of patients with non-Hodgkin B-cell lymphomas. Expert Opin. Investig. Drugs 2018, 27, 601–611. [Google Scholar] [CrossRef] [PubMed]
  201. Ribera, J.M.; Genescà, E.; Ribera, J. Bispecific T-cell engaging antibodies in B-cell precursor acute lymphoblastic leukemias: Focus on blinatumomab. Ther. Adv. Hematol. 2020, 11, 2040620720919632. [Google Scholar] [CrossRef]
  202. Xu, B. Loncastuximab tesirine: An effective therapy for relapsed or refractory diffuse large B-cell lymphoma. Eur. J. Clin. Pharmacol. 2022, 78, 707–719. [Google Scholar] [CrossRef]
  203. Sesques, P.; Ferrant, E.; Safar, V.; Wallet, F.; Tordo, J.; Dhomps, A.; Karlin, L.; Brisou, G.; Vercasson, M.; Hospital-Gustem, C.; et al. Commercial anti-CD19 CAR T cell therapy for patients with relapsed/refractory aggressive B cell lymphoma in a European center. Am. J. Hematol. 2020, 95, 1324–1333. [Google Scholar] [CrossRef]
  204. Sharma, P.; King, G.T.; Shinde, S.S.; Purev, E.; Jimeno, A. Axicabtagene ciloleucel for the treatment of relapsed/refractory B-cell non-Hodgkin’s lymphomas. Drugs Today 2018, 54, 187–198. [Google Scholar] [CrossRef]
  205. Abramson, J.S. Anti-CD19 CAR T-Cell Therapy for B-Cell Non-Hodgkin Lymphoma. Transfus. Med. Rev. 2020, 34, 29–33. [Google Scholar] [CrossRef]
  206. Dhakal, B.; Hari, P.N.; Usmani, S.Z.; Hamadani, M. Chimeric antigen receptor T cell therapy in multiple myeloma: Promise and challenges. Bone Marrow Transplant. 2021, 56, 9–19. [Google Scholar] [CrossRef] [PubMed]
  207. Rambaldi, A.; Ribera, J.M.; Kantarjian, H.M.; Dombret, H.; Ottmann, O.G.; Stein, A.S.; Tuglus, C.A.; Zhao, X.; Kim, C.; Martinelli, G. Blinatumomab compared with standard of care for the treatment of adult patients with relapsed/refractory Philadelphia chromosome-positive B-precursor acute lymphoblastic leukemia. Cancer 2020, 126, 304–310. [Google Scholar] [CrossRef]
  208. Horton, H.M.; Bernett, M.J.; Pong, E.; Peipp, M.; Karki, S.; Chu, S.Y.; Richards, J.O.; Vostiar, I.; Joyce, P.F.; Repp, R.; et al. Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia. Cancer Res. 2008, 68, 8049–8057. [Google Scholar] [CrossRef] [Green Version]
  209. Awan, F.T.; Lapalombella, R.; Trotta, R.; Butchar, J.P.; Yu, B.; Benson, D.M., Jr.; Roda, J.M.; Cheney, C.; Mo, X.; Lehman, A.; et al. CD19 targeting of chronic lymphocytic leukemia with a novel Fc-domain-engineered monoclonal antibody. Blood 2010, 115, 1204–1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Caimi, P.F.; Ai, W.; Alderuccio, J.P.; Ardeshna, K.M.; Hamadani, M.; Hess, B.; Kahl, B.S.; Radford, J.; Solh, M.; Stathis, A.; et al. Loncastuximab tesirine in relapsed or refractory diffuse large B-cell lymphoma (LOTIS-2): A multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2021, 22, 790–800. [Google Scholar] [CrossRef]
  211. Halford, Z.; Anderson, M.K.; Bennett, L.L. Axicabtagene Ciloleucel: Clinical Data for the Use of CAR T-cell Therapy in Relapsed and Refractory Large B-cell Lymphoma. Ann. Pharmacother. 2021, 55, 390–405. [Google Scholar] [CrossRef] [PubMed]
  212. Jain, M.D.; Bachmeier, C.A.; Phuoc, V.H.; Chavez, J.C. Axicabtagene ciloleucel (KTE-C19), an anti-CD19 CAR T therapy for the treatment of relapsed/refractory aggressive B-cell non-Hodgkin’s lymphoma. Ther. Clin. Risk Manag. 2018, 14, 1007–1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Vairy, S.; Garcia, J.L.; Teira, P.; Bittencourt, H. CTL019 (tisagenlecleucel): CAR-T therapy for relapsed and refractory B-cell acute lymphoblastic leukemia. Drug Des. Dev. Ther. 2018, 12, 3885–3898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Kamdar, M.; Solomon, S.R.; Arnason, J.; Johnston, P.B.; Glass, B.; Bachanova, V.; Ibrahimi, S.; Mielke, S.; Mutsaers, P.; Hernandez-Ilizaliturri, F.; et al. Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): Results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet 2022, 399, 2294–2308. [Google Scholar]
  215. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef]
  216. Laetsch, T.W.; Maude, S.L.; Balduzzi, A.; Rives, S.; Bittencourt, H.; Boyer, M.W.; Buechner, J.; De Moerloose, B.; Qayed, M.; Phillips, C.L.; et al. Tisagenlecleucel in pediatric and young adult patients with Down syndrome-associated relapsed/refractory acute lymphoblastic leukemia. Leukemia 2022, 36, 1508–1515. [Google Scholar] [CrossRef]
  217. Moskop, A.; Pommert, L.; Baggott, C.; Prabhu, S.; Pacenta, H.L.; Phillips, C.L.; Rossoff, J.; Stefanski, H.E.; Talano, J.A.; Margossian, S.P.; et al. Real-world use of tisagenlecleucel in infant acute lymphoblastic leukemia. Blood Adv. 2022, 6, 4251–4255. [Google Scholar] [CrossRef]
  218. Kwon, M.; Iacoboni, G.; Reguera, J.L.; Corral, L.L.; Morales, R.H.; Ortiz-Maldonado, V.; Guerreiro, M.; Caballero, A.C.; Domínguez, M.L.G.; Pina, J.M.S.; et al. Axicabtagene ciloleucel compared to tisagenlecleucel for the treatment of aggressive B-cell lymphoma. Haematologica 2022. Online ahead of print. [Google Scholar] [CrossRef]
  219. Ghione, P.; Palomba, M.L.; Patel, A.; Bobillo, S.; Deighton, K.; Jacobson, C.A.; Nahas, M.; Hatswell, A.J.; Jung, A.S.; Kanters, S.; et al. Comparative effectiveness of ZUMA-5 (axi-cel) vs. SCHOLAR-5 external control in relapsed/refractory follicular lymphoma. Blood 2022, 140, 851–860. [Google Scholar] [CrossRef] [PubMed]
  220. Locke, F.L.; Miklos, D.B.; Jacobson, C.A.; Perales, M.A.; Kersten, M.J.; Oluwole, O.O.; Ghobadi, A.; Rapoport, A.P.; McGuirk, J.; Pagel, J.M.; et al. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N. Engl. J. Med. 2022, 386, 640–654. [Google Scholar] [CrossRef]
  221. Siddiqi, T.; Soumerai, J.D.; Dorritie, K.A.; Stephens, D.M.; Riedell, P.A.; Arnason, J.; Kipps, T.J.; Gillenwater, H.H.; Gong, L.; Yang, L.; et al. Phase 1 TRANSCEND CLL 004 study of lisocabtagene maraleucel in patients with relapsed/refractory CLL or SLL. Blood 2022, 139, 1794–1806. [Google Scholar] [CrossRef]
  222. Chavez, J.C.; Locke, F.L. CAR T cell therapy for B-cell lymphomas. Best Pract. Res. Clin. Haematol. 2018, 31, 135–146. [Google Scholar] [CrossRef]
  223. Shah, N.N.; Maatman, T.; Hari, P.; Johnson, B. Multi Targeted CAR-T Cell Therapies for B-Cell Malignancies. Front. Oncol. 2019, 9, 146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Messéant, O.; Houot, R. CAR-T cells in lymphomas: Current and evolving role. Bull. Cancer 2021, 108, S28–S39. [Google Scholar] [CrossRef]
  225. Martyniszyn, A.; Krahl, A.C.; André, M.C.; Hombach, A.A.; Abken, H. CD20-CD19 Bispecific CAR T Cells for the Treatment of B-Cell Malignancies. Hum. Gene Ther. 2017, 28, 1147–1157. [Google Scholar] [CrossRef] [PubMed]
  226. Tong, C.; Zhang, Y.; Liu, Y.; Ji, X.; Zhang, W.; Guo, Y.; Han, X.; Ti, D.; Dai, H.; Wang, C.; et al. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B-cell lymphoma. Blood 2020, 136, 1632–1644. [Google Scholar] [CrossRef] [PubMed]
  227. Zhang, C.; Hu, Y.; Shi, C. Targeting Natural Killer Cells for Tumor Immunotherapy. Front. Immunol. 2020, 11, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Heipertz, E.L.; Zynda, E.R.; Stav-Noraas, T.E.; Hungler, A.D.; Boucher, S.E.; Kaur, N.; Vemuri, M.C. Current Perspectives on “Off-The-Shelf” Allogeneic NK and CAR-NK Cell Therapies. Front. Immunol. 2021, 12, 732135. [Google Scholar] [CrossRef]
  229. Kantarjian, H.M.; Su, Y.; Jabbour, E.J.; Bhattacharyya, H.; Yan, E.; Cappelleri, J.C.; Marks, D.I. Patient-reported outcomes from a phase 3 randomized controlled trial of inotuzumab ozogamicin versus standard therapy for relapsed/refractory acute lymphoblastic leukemia. Cancer 2018, 124, 2151–2160. [Google Scholar] [CrossRef]
  230. Jabbour, E.; Stelljes, M.; Advani, A.S.; DeAngelo, D.J.; Wang, T.; Neuhof, A.; Vandendries, E.; Kantarjian, H.M. Time from randomization to first subsequent induction/salvage therapy (ST) in patients with relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL) treated with inotuzumab ozogamicin (InO) in the phase 3 INO-VATE trial. J. Clin. Oncol. 2019, 373, 7025. [Google Scholar] [CrossRef]
  231. Shah, N.N.; Bhojwani, D.; August, K.; Baruchel, A.; Bertrand, Y.; Boklan, J.; Dalla-Pozza, L.; Dennis, R.; Hijiya, N.; Locatelli, F.; et al. Results from an international phase 2 study of the anti-CD22 immunotoxin moxetumomab pasudotox in relapsed or refractory childhood B-lineage acute lymphoblastic leukemia. Pediatr Blood Cancer 2020, 67, e28112. [Google Scholar] [CrossRef]
  232. Contreras, C.F.; Higham, C.S.; Behnert, A.; Kim, K.; Stieglitz, E.; Tasian, S.K. Clinical utilization of blinatumomab and inotuzumab immunotherapy in children with relapsed or refractory B-acute lymphoblastic leukemia. Pediatr. Blood Cancer 2021, 68, e28718. [Google Scholar] [CrossRef] [PubMed]
  233. Kreitman, R.J.; Pastan, I. Antibody fusion proteins: Anti-CD22 recombinant immunotoxin moxetumomab pasudotox. Clin. Cancer Res. 2011, 17, 6398–6405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Jabbour, E.; Sasaki, K.; Ravandi, F.; Huang, X.; Short, N.J.; Khouri, M.; Kebriaei, P.; Burger, J.; Khoury, J.; Jorgensen, J.; et al. Chemoimmunotherapy with inotuzumab ozogamicin combined with mini-hyper-CVD, with or without blinatumomab, is highly effective in patients with Philadelphia chromosome-negative acute lymphoblastic leukemia in first salvage. Cancer 2018, 124, 4044–4055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Pennesi, E.; Michels, N.; Brivio, E.; van der Velden, V.H.J.; Jiang, Y.; Thano, A.; Ammerlaan, A.J.C.; Boer, J.M.; Beverloo, H.B.; Sleight, B.; et al. Inotuzumab ozogamicin as single agent in pediatric patients with relapsed and refractory acute lymphoblastic leukemia: Results from a phase II trial. Leukemia 2022, 36, 1516–1524. [Google Scholar] [CrossRef] [PubMed]
  236. Advani, A.; Coiffier, B.; Czuczman, M.S.; Dreyling, M.; Foran, J.; Gine, E.; Gisselbrecht, C.; Ketterer, N.; Nasta, S.; Rohatiner, A.; et al. Safety, pharmacokinetics, and preliminary clinical activity of inotuzumab ozogamicin, a novel immunoconjugate for the treatment of B-cell non-Hodgkin’s lymphoma: Results of a phase I study. J. Clin. Oncol. 2010, 28, 2085–2093. [Google Scholar] [CrossRef] [PubMed]
  237. Ogura, M.; Tobinai, K.; Hatake, K.; Uchida, T.; Kasai, M.; Oyama, T.; Suzuki, T.; Kobayashi, Y.; Watanabe, T.; Azuma, T.; et al. Phase I study of inotuzumab ozogamicin (CMC-544) in Japanese patients with follicular lymphoma pretreated with rituximab-based therapy. Cancer Sci. 2010, 101, 1840–1845. [Google Scholar] [CrossRef]
  238. Fayad, L.; Offner, F.; Smith, M.R.; Verhoef, G.; Johnson, P.; Kaufman, J.L.; Rohatiner, A.; Advani, A.; Foran, J.; Hess, G.; et al. Safety and clinical activity of a combination therapy comprising two antibody-based targeting agents for the treatment of non-Hodgkin lymphoma: Results of a phase I/II study evaluating the immunoconjugate inotuzumab ozogamicin with rituximab. J. Clin. Oncol. 2013, 31, 573–583. [Google Scholar] [CrossRef]
  239. Short, N.J.; Kantarjian, H.; Jabbour, E.; Cortes, J.E.; Thomas, D.A.; Rytting, M.E.; Daver, N.; Alvarado, Y.; Konopleva, M.; Kebriaei, P.; et al. A phase I study of moxetumomab pasudotox in adults with relapsed or refractory B-cell acute lymphoblastic leukaemia. Br. J. Haematol. 2018, 182, 442–444. [Google Scholar] [CrossRef] [Green Version]
  240. Haso, W.; Lee, D.W.; Shah, N.N.; Stetler-Stevenson, M.; Yuan, C.M.; Pastan, I.H.; Dimitrov, D.S.; Morgan, R.A.; FitzGerald, D.J.; Barrett, D.M.; et al. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood 2013, 121, 1165–1174. [Google Scholar] [CrossRef] [Green Version]
  241. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet 2015, 385, 517–528. [Google Scholar] [CrossRef]
  242. Shalabi, H.; Qin, H.; Su, A.; Yates, B.; Wolters, P.L.; Steinberg, S.M.; Ligon, J.A.; Silbert, S.; DéDé, K.; Benzaoui, M.; et al. CD19/22 CAR T-cells in Children and Young Adults with B-ALL: Phase I Results and Development of a Novel Bicistronic CAR. Blood 2022, 140, 451–463. [Google Scholar] [CrossRef]
  243. Usmani, S.Z.; Quach, H.; Mateos, M.V.; Landgren, O.; Leleu, X.; Siegel, D.; Weisel, K.; Gavriatopoulou, M.; Oriol, A.; Rabin, N.; et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): Updated outcomes from a randomised, multicentre, open-label, phase 3 study. Lancet Oncol. 2022, 23, 65–76. [Google Scholar] [CrossRef]
  244. Dimopoulos, M.; Bringhen, S.; Anttila, P.; Capra, M.; Cavo, M.; Cole, C.; Gasparetto, C.; Hungria, V.; Jenner, M.; Vorobyev, V.; et al. Isatuximab as monotherapy and combined with dexamethasone in patients with relapsed/refractory multiple myeloma. Blood 2021, 137, 1154–1165. [Google Scholar] [CrossRef]
  245. Moreau, P.; Dimopoulos, M.A.; Mikhael, J.; Yong, K.; Capra, M.; Facon, T.; Hajek, R.; Špička, I.; Baker, R.; Kim, K.; et al. Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): A multicentre, open-label, randomised phase 3 trial. Lancet 2021, 397, 2361–2371. [Google Scholar] [CrossRef]
  246. Richardson, P.G.; Harrison, S.J.; Bringhen, S.; Schjesvold, F.; Yong, K.; Campana, F.; Le-Guennec, S.; Macé, S.; Dimopoulos, M.A. Isatuximab for relapsed/refractory multiple myeloma: Review of key subgroup analyses from the Phase III ICARIA-MM study. Future Oncol. 2021, 17, 4797–4812. [Google Scholar] [CrossRef] [PubMed]
  247. Calabretta, E.; Carlo-Stella, C. The Many Facets of CD38 in Lymphoma: From Tumor-Microenvironment Cell Interactions to Acquired Resistance to Immunotherapy. Cells 2020, 9, 802. [Google Scholar] [CrossRef] [Green Version]
  248. Gozzetti, A.; Ciofini, S.; Simoncelli, M.; Santoni, A.; Pacelli, P.; Raspadori, D.; Bocchia, M. Anti CD38 monoclonal antibodies for multiple myeloma treatment. Hum. Vaccines Immunother. 2022, 18, 2052658. [Google Scholar] [CrossRef]
  249. Jiang, H.; Acharya, C.; An, G.; Zhong, M.; Feng, X.; Wang, L.; Dasilva, N.; Song, Z.; Yang, G.; Adrian, F.; et al. SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia 2016, 30, 399–408. [Google Scholar] [CrossRef] [PubMed]
  250. Franssen, L.E.; Stege, C.A.M.; Zweegman, S.; van de Donk, N.; Nijhof, I.S. Resistance Mechanisms Towards CD38-Directed Antibody Therapy in Multiple Myeloma. J. Clin. Med. 2020, 9, 1195. [Google Scholar] [CrossRef] [Green Version]
  251. Yang, Y.; Li, Y.; Gu, H.; Dong, M.; Cai, Z. Emerging agents and regimens for multiple myeloma. J. Hematol. Oncol. 2020, 13, 150. [Google Scholar] [CrossRef]
  252. Usmani, S.Z.; Karanes, C.; Bensinger, W.I.; D’Souza, A.; Raje, N.; Tuchman, S.A.; Sborov, D.; Laubach, J.P.; Bianchi, G.; Kanagavel, D.; et al. Final results of a phase 1b study of isatuximab short-duration fixed-volume infusion combination therapy for relapsed/refractory multiple myeloma. Leukemia 2021, 35, 3526–3533. [Google Scholar] [CrossRef] [PubMed]
  253. Voorhees, P.M.; Kaufman, J.L.; Laubach, J.; Sborov, D.W.; Reeves, B.; Rodriguez, C.; Chari, A.; Silbermann, R.; Costa, L.J.; Anderson, L.D., Jr.; et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: The GRIFFIN trial. Blood 2020, 136, 936–945. [Google Scholar] [CrossRef] [PubMed]
  254. Matas-Céspedes, A.; Vidal-Crespo, A.; Rodriguez, V.; Villamor, N.; Delgado, J.; Giné, E.; Roca-Ho, H.; Menéndez, P.; Campo, E.; López-Guillermo, A.; et al. The Human CD38 Monoclonal Antibody Daratumumab Shows Antitumor Activity and Hampers Leukemia-Microenvironment Interactions in Chronic Lymphocytic Leukemia. Clin. Cancer Res. 2017, 23, 1493–1505. [Google Scholar] [CrossRef] [Green Version]
  255. Manna, A.; Aulakh, S.; Jani, P.; Ahmed, S.; Akhtar, S.; Coignet, M.; Heckman, M.; Meghji, Z.; Bhatia, K.; Sharma, A.; et al. Targeting CD38 Enhances the Antileukemic Activity of Ibrutinib in Chronic Lymphocytic Leukemia. Clin. Cancer Res. 2019, 25, 3974–3985. [Google Scholar] [CrossRef] [Green Version]
  256. Wang, A.; Song, Z.; Zheng, G.; Nicolazzi, C.; Fromm, J.R.; Shehu, E.; Srinivasan, S.; Chen, X.; Zhu, C.; Blondel, M.C.; et al. Evaluation of Preclinical Activity of Isatuximab in Patients with Acute Lymphoblastic Leukemia. Mol. Cancer Ther. 2021, 20, 1916–1925. [Google Scholar] [CrossRef] [PubMed]
  257. Baruchel, A.; Abrahamsson, J.; Bertrand, Y.; Gonzalez, O.; Nysom, K.; Quinones, W.; Rizzari, C.; Buechner, J.; Cesaro, S.; Duarte, J.; et al. Isatuximab in Combination with Chemotherapy in Pediatric Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia or Acute Myeloid Leukemia (ISAKIDS): Interim Analysis. Blood 2021, 138, 516. [Google Scholar] [CrossRef]
  258. Hashmi, H.; Husnain, M.; Khan, A.; Usmani, S.Z. CD38-Directed Therapies for Management of Multiple Myeloma. Immunotargets Ther. 2021, 10, 201–211. [Google Scholar] [CrossRef]
  259. Abramson, H.N. Emerging Monoclonal Antibodies for the Treatment of Multiple Myeloma; IntechOpen: London, UK, 2020; pp. 1–20. [Google Scholar]
  260. Raab, M.S.; Engelhardt, M.; Blank, A.; Goldschmidt, H.; Agis, H.; Blau, I.W.; Einsele, H.; Ferstl, B.; Schub, N.; Röllig, C.; et al. MOR202, a novel anti-CD38 monoclonal antibody, in patients with relapsed or refractory multiple myeloma: A first-in-human, multicentre, phase 1-2a trial. Lancet Haematol. 2020, 7, e381–e394. [Google Scholar] [CrossRef]
  261. Krishnan, A.Y.; Patel, K.K.; Hari, P.; Jagannath, S.; Niesvizky, R.; Silbermann, R.W.; Berg, D.T.; Li, Q.; Allikmets, K.; Stockerl-Goldstein, K. A phase Ib study of TAK-079, an investigational anti-CD38 monoclonal antibody (mAb) in patients with relapsed/refractory multiple myeloma (RRMM): Preliminary results. J. Clin. Oncol. 2020, 38, 8539. [Google Scholar] [CrossRef]
  262. Zuch de Zafra, C.L.; Fajardo, F.; Zhong, W.; Bernett, M.J.; Muchhal, U.S.; Moore, G.L.; Stevens, J.; Case, R.; Pearson, J.T.; Liu, S.; et al. Targeting Multiple Myeloma with AMG 424, a Novel Anti-CD38/CD3 Bispecific T-cell-recruiting Antibody Optimized for Cytotoxicity and Cytokine Release. Clin. Cancer Res. 2019, 25, 3921–3933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Doucey, M.A.; Estoppey, C.; Stutz, C.; Croset, A.; Laurendon, A.; Monney, T.; Pluess, M.; Ries-Fecourt, C.; Macoin, J.; Turrini, R.; et al. ISB 1342: A first-in-class CD38 T cell engager for the treatment of relapsed refractory multiple myeloma. J. Clin. Oncol. 2021, 39, 8044. [Google Scholar] [CrossRef]
  264. Guo, Y.; Feng, K.; Tong, C.; Jia, H.; Liu, Y.; Wang, Y.; Ti, D.; Yang, Q.; Wu, Z.; Han, W. Efficiency and side effects of anti-CD38 CAR T cells in an adult patient with relapsed B-ALL after failure of bi-specific CD19/CD22 CAR T cell treatment. Cell. Mol. Immunol. 2020, 17, 430–432. [Google Scholar] [CrossRef]
  265. Duggal, R.; Santara, S.S.; Gordon, M.; Kilgallon, A.; Hermanson, D.; Childs, R.W.; O’Dwyer, M.E. Promising Preliminary Activity of Optimized Affinity, CD38 CAR NK Cells Generated Using a Non-Viral Engineering Approach in Gene Edited Cord Blood Derived NK Cells for the Treatment of Multiple Myeloma. Blood 2021, 138, 4793. [Google Scholar] [CrossRef]
  266. Stikvoort, A.; van der Schans, J.; Sarkar, S.; Poels, R.; Ruiter, R.; Naik, J.; Yuan, H.; de Bruijn, J.D.; van de Donk, N.W.; Zweegman, S.; et al. CD38-specific Chimeric Antigen Receptor Expressing Natural Killer KHYG-1 Cells: A Proof of Concept for an “Off the Shelf” Therapy for Multiple Myeloma. HemaSphere 2021, 5, e596. [Google Scholar] [CrossRef]
  267. Cho, S.F.; Anderson, K.C.; Tai, Y.T. Targeting B Cell Maturation Antigen (BCMA) in Multiple Myeloma: Potential Uses of BCMA-Based Immunotherapy. Front. Immunol. 2018, 9, 1821. [Google Scholar] [CrossRef] [Green Version]
  268. Zhou, X.; Mulazzani, M.; von Mücke-Heim, I.A.; Langer, S.; Zhang, W.; Ishikawa-Ankerhold, H.; Dreyling, M.; Straube, A.; von Baumgarten, L. The Role of BAFF-R Signaling in the Growth of Primary Central Nervous System Lymphoma. Front. Oncol. 2020, 10, 682. [Google Scholar] [CrossRef]
  269. Sanchez, L.; Dardac, A.; Madduri, D.; Richard, S.; Richter, J. B-cell maturation antigen (BCMA) in multiple myeloma: The new frontier of targeted therapies. Ther. Adv. Hematol. 2021, 12, 2040620721989585. [Google Scholar] [CrossRef]
  270. Lancman, G.; Richter, J.; Chari, A. Bispecifics, trispecifics, and other novel immune treatments in myeloma. Hematol. Am. Soc. Hematol. Educ. Program 2020, 2020, 264–271. [Google Scholar] [CrossRef]
  271. Strassl, I.; Schreder, M.; Steiner, N.; Rudzki, J.; Agis, H.; Künz, T.; Müser, N.; Willenbacher, W.; Petzer, A.; Neumeister, P.; et al. The Agony of Choice-Where to Place the Wave of BCMA-Targeted Therapies in the Multiple Myeloma Treatment Puzzle in 2022 and Beyond. Cancers 2021, 13, 4701. [Google Scholar] [CrossRef]
  272. Sheikh, S.; Lebel, E.; Trudel, S. Belantamab mafodotin in the treatment of relapsed or refractory multiple myeloma. Future Oncol. 2020, 16, 2783–2798. [Google Scholar] [CrossRef] [PubMed]
  273. Trudel, S.; Nooka, A.; Fecteau, D.; Talekar, M.; Jewell, R.C.; Williams, D.; Evans, J.; Opalinska, J. DREAMM 4: A phase I/II single-arm open-label study to explore safety and clinical activity of belantamab mafodotin (GSK2857916) administered in combination with pembrolizumab in patients with relapsed/refractory multiple myeloma (RRMM). Ann. Oncol. 2019, 30, v447. [Google Scholar] [CrossRef]
  274. Xing, L.; Lin, L.; Yu, T.; Li, Y.; Cho, S.F.; Liu, J.; Wen, K.; Hsieh, P.A.; Kinneer, K.; Munshi, N.; et al. A novel BCMA PBD-ADC with ATM/ATR/WEE1 inhibitors or bortezomib induce synergistic lethality in multiple myeloma. Leukemia 2020, 34, 2150–2162. [Google Scholar] [CrossRef]
  275. Kumar, S.; Migkou, M.; Bhutani, M.; Spencer, A.; Ailawadhi, S.; Kalff, A.; Walcott, F.; Pore, N.; Gibson, D.; Wang, F.; et al. Phase 1, First-in-Human Study of MEDI2228, a BCMA-Targeted ADC in Patients with Relapsed/Refractory Multiple Myeloma. Blood 2020, 136, 26–27. [Google Scholar] [CrossRef]
  276. Trudel, S.; Lendvai, N.; Popat, R.; Voorhees, P.M.; Reeves, B.; Libby, E.N.; Richardson, P.G.; Hoos, A.; Gupta, I.; Bragulat, V.; et al. Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: An update on safety and efficacy from dose expansion phase I study. Blood Cancer J. 2019, 9, 37. [Google Scholar] [CrossRef] [Green Version]
  277. Lee, H.C.; Raje, N.S.; Landgren, O.; Upreti, V.V.; Wang, J.; Avilion, A.A.; Hu, X.; Rasmussen, E.; Ngarmchamnanrith, G.; Fujii, H.; et al. Phase 1 study of the anti-BCMA antibody-drug conjugate AMG 224 in patients with relapsed/refractory multiple myeloma. Leukemia 2021, 35, 255–258. [Google Scholar] [CrossRef]
  278. Usmani, S.Z.; Garfall, A.L.; van de Donk, N.; Nahi, H.; San-Miguel, J.F.; Oriol, A.; Rosinol, L.; Chari, A.; Bhutani, M.; Karlin, L.; et al. Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, in patients with relapsed or refractory multiple myeloma (MajesTEC-1): A multicentre, open-label, single-arm, phase 1 study. Lancet 2021, 398, 665–674. [Google Scholar] [CrossRef]
  279. Munshi, N.C.; Anderson, L.D., Jr.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716. [Google Scholar] [CrossRef]
  280. Costa, L.J.; Lin, Y.; Cornell, R.F.; Martin, T.; Chhabra, S.; Usmani, S.Z.; Jagannath, S.; Callander, N.S.; Berdeja, J.G.; Kang, Y.; et al. Comparison of Cilta-cel, an Anti-BCMA CAR-T Cell Therapy, Versus Conventional Treatment in Patients With Relapsed/Refractory Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2022, 22, 326–335. [Google Scholar] [CrossRef] [PubMed]
  281. Tang, Y.; Yin, H.; Zhao, X.; Jin, D.; Liang, Y.; Xiong, T.; Li, L.; Tang, W.; Zhang, J.; Liu, M.; et al. High efficacy and safety of CD38 and BCMA bispecific CAR-T in relapsed or refractory multiple myeloma. J. Exp. Clin. Cancer Res. 2022, 41, 2. [Google Scholar] [CrossRef] [PubMed]
  282. Ng, Y.Y.; Du, Z.; Zhang, X.; Chng, W.J.; Wang, S. CXCR4 and anti-BCMA CAR co-modified natural killer cells suppress multiple myeloma progression in a xenograft mouse model. Cancer Gene Ther. 2022, 29, 475–483. [Google Scholar] [CrossRef]
  283. Ding, W.; LaPlant, B.R.; Call, T.G.; Parikh, S.A.; Leis, J.F.; He, R.; Shanafelt, T.D.; Sinha, S.; Le-Rademacher, J.; Feldman, A.L.; et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017, 129, 3419–3427. [Google Scholar] [CrossRef]
  284. Sandhu, K.S.; Huynh-Tran, Q.; Cooper, E.E.; Palmer, J.; Tsai, N.C.; Thomas, S.; Robbins, M.; Aribi, A.; Salhotra, A.; Mei, M.; et al. ALL-440: Promising Safety and Efficacy Results from an Ongoing Phase 1/2 Study of Pembrolizumab in Combination with Blinatumomab in Patients (pts) with Relapsed or Refractory (R/R) Acute Lymphoblastic Leukemia (ALL). Clin. Lymphoma Leukemia 2021, 21, 276. [Google Scholar] [CrossRef]
  285. Carlo-Stella, C.; Zinzani, P.L.L.; Sureda, A.; Araújo, L.F.; Casasnovas, O.; Carpio, C.; Yeh, S.P.; Bouabdallah, K.; Cartron, G.; Kim, W.S.; et al. A Phase 1/2, Open-Label, Multicenter Study of Isatuximab in Combination with Cemiplimab in Patients with Lymphoma. Blood 2021, 138, 4362. [Google Scholar] [CrossRef]
  286. Jacobson, C.S.J. End of Phase 1 Results from Zuma-6: Axicabtagene Ciloleucel (Axi-Cel) in Combination with Atezolizumab for the Treatment of Patients with Refractory Diffuse Large B Cell Lymphoma. Blood 2018, 132, 4192. [Google Scholar] [CrossRef]
  287. Treon, S.P.; Meid, K.; Hunter, Z.R.; Flynn, C.A.; Sarosiek, S.R.; Leventoff, C.R.; White, T.P.; Cao, Y.; Roccaro, A.M.; Sacco, A.; et al. Phase 1 study of ibrutinib and the CXCR4 antagonist ulocuplumab in CXCR4-mutated Waldenström macroglobulinemia. Blood 2021, 138, 1535–1539. [Google Scholar] [CrossRef]
  288. Schjesvold, F.; Robak, P.; Pour, L.; Aschan, J.; Sonneveld, P. OCEAN: A randomized Phase III study of melflufen + dexamethasone to treat relapsed refractory multiple myeloma. Future Oncol. 2020, 16, 631–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  289. Kolstad, A.; Illidge, T.; Bolstad, N.; Spetalen, S.; Madsbu, U.; Stokke, C.; Blakkisrud, J.; Løndalen, A.; O’Rourke, N.; Beasley, M.; et al. Phase 1/2a study of 177Lu-lilotomab satetraxetan in relapsed/refractory indolent non-Hodgkin lymphoma. Blood Adv. 2020, 4, 4091–4101. [Google Scholar] [CrossRef] [PubMed]
  290. Wang, C.M.; Wu, Z.Q.; Wang, Y.; Guo, Y.L.; Dai, H.R.; Wang, X.H.; Li, X.; Zhang, Y.J.; Zhang, W.Y.; Chen, M.X.; et al. Autologous T Cells Expressing CD30 Chimeric Antigen Receptors for Relapsed or Refractory Hodgkin Lymphoma: An Open-Label Phase I Trial. Clin. Cancer Res. 2017, 23, 1156–1166. [Google Scholar] [CrossRef] [Green Version]
  291. Ramos, C.A.; Ballard, B.; Zhang, H.; Dakhova, O.; Gee, A.P.; Mei, Z.; Bilgi, M.; Wu, M.F.; Liu, H.; Grilley, B.; et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J. Clin. Investig. 2017, 127, 3462–3471. [Google Scholar] [CrossRef] [Green Version]
  292. Advani, R.H.; Flinn, I.; Popplewell, L.; Forero-Torres, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.P.; Tran, T.; Lynn, J.; Chen, J.Y.; et al. Activity and tolerabilty of the first-in-class anti-CD47 antibody Hu5F9-G4 with rituximab tolerated in relapsed/refractory non-Hodgkin lymphoma: Initial phase 1b/2 results. J. Clin. Oncol. 2018, 36, 7504. [Google Scholar] [CrossRef]
  293. Chauchet, X.; Cons, L.; Chatel, L.; Daubeuf, B.; Didelot, G.; Moine, V.; Chollet, D.; Malinge, P.; Pontini, G.; Masternak, K.; et al. CD47xCD19 bispecific antibody triggers recruitment and activation of innate immune effector cells in a B-cell lymphoma xenograft model. Exp. Hematol. Oncol. 2022, 11, 26. [Google Scholar] [CrossRef]
  294. Krishnan, A.Y.; Shah, N.; Spira, A.I.; Kaufman, J.L.; Niesvizky, R.; Shah, N.N.; Burke, J.M.; Popplewell, L.; Martin, T.G.; Cheung, J.; et al. A phase 1 open-label, safety, pharmacokinetic, and preliminary efficacy study of STRO-001, an anti-CD74 antibody drug conjugate, in patients with advanced B-cell malignancies. J. Clin. Oncol. 2018, 36, 7586. [Google Scholar] [CrossRef]
  295. Cohen, A.D.; Harrison, S.J.; Krishnan, A.; Fonseca, R.; Forsberg, P.A.; Spencer, A.; Berdeja, J.G.; Laubach, J.P.; Li, M.; Choeurng, V.; et al. Initial clinical activity and safety of BFCR4350A, a FcRH5/CD3 T-cell-engaging bispecific antibody, in relapsed/refractory multiple myeloma. Blood 2020, 136, 42–43. [Google Scholar] [CrossRef]
  296. Berdeja, J.G.; Krishnan, A.Y.; Oriol, A.; van de Donk, N.W.; Rodríguez-Otero, P.; Askari, E.; Mateos, M.V.; Minnema, M.C.; Costa, L.J.; Verona, R.; et al. Updated results of a phase 1, first-in-human study of talquetamab, a G protein-coupled receptor family C group 5 member D (GPRC5D) × CD3 bispecific antibody, in relapsed/refractory multiple myeloma (MM). J. Clin. Oncol. 2021, 39, 8008. [Google Scholar] [CrossRef]
  297. Cui, B.; Ghia, E.M.; Chen, L.; Rassenti, L.Z.; DeBoever, C.; Widhopf, G.F., 2nd; Yu, J.; Neuberg, D.S.; Wierda, W.G.; Rai, K.R.; et al. High-level ROR1 associates with accelerated disease progression in chronic lymphocytic leukemia. Blood 2016, 128, 2931–2940. [Google Scholar] [CrossRef] [Green Version]
  298. Fukuda, T.; Chen, L.; Endo, T.; Tang, L.; Lu, D.; Castro, J.E.; Widhopf, G.F., 2nd; Rassenti, L.Z.; Cantwell, M.J.; Prussak, C.E.; et al. Antisera induced by infusions of autologous Ad-CD154-leukemia B cells identify ROR1 as an oncofetal antigen and receptor for Wnt5a. Proc. Natl. Acad. Sci. USA 2008, 105, 3047–3052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  299. Yu, J.; Chen, L.; Cui, B.; Widhopf, G.F., 2nd; Shen, Z.; Wu, R.; Zhang, L.; Zhang, S.; Briggs, S.P.; Kipps, T.J. Wnt5a induces ROR1/ROR2 heterooligomerization to enhance leukemia chemotaxis and proliferation. J. Clin. Investig. 2016, 126, 585–598. [Google Scholar] [CrossRef]
  300. Ghia, E.M.; Rassenti, L.Z.; Choi, M.Y.; Quijada-Álamo, M.; Chu, E.; Widhopf, G.F., 2nd; Kipps, T.J. High expression level of ROR1 and ROR1-signaling associates with venetoclax resistance in chronic lymphocytic leukemia. Leukemia 2022, 36, 1609–1618. [Google Scholar] [CrossRef]
  301. Choi, M.Y.; Widhopf, G.F., 2nd; Ghia, E.M.; Kidwell, R.L.; Hasan, M.K.; Yu, J.; Rassenti, L.Z.; Chen, L.; Chen, Y.; Pittman, E.; et al. Phase I Trial: Cirmtuzumab Inhibits ROR1 Signaling and Stemness Signatures in Patients with Chronic Lymphocytic Leukemia. Cell Stem Cell 2018, 22, 951–959.e3. [Google Scholar] [CrossRef] [Green Version]
  302. Beenen, A.C.; Sauerer, T.; Schaft, N.; Dörrie, J. Beyond Cancer: Regulation and Function of PD-L1 in Health and Immune-Related Diseases. Int. J. Mol. Sci. 2022, 23, 8599. [Google Scholar] [CrossRef] [PubMed]
  303. Zhao, X.; Bao, Y.; Meng, B.; Xu, Z.; Li, S.; Wang, X.; Hou, R.; Ma, W.; Liu, D.; Zheng, J.; et al. From rough to precise: PD-L1 evaluation for predicting the efficacy of PD-1/PD-L1 blockades. Front. Immunol. 2022, 13, 920021. [Google Scholar] [CrossRef] [PubMed]
  304. Wang, Y.; Zhang, H.; Liu, C.; Wang, Z.; Wu, W.; Zhang, N.; Zhang, L.; Hu, J.; Luo, P.; Zhang, J.; et al. Immune checkpoint modulators in cancer immunotherapy: Recent advances and emerging concepts. J. Hematol. Oncol. 2022, 15, 111. [Google Scholar] [CrossRef] [PubMed]
  305. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
  306. Boyiadzis, M.M.; Kirkwood, J.M.; Marshall, J.L.; Pritchard, C.C.; Azad, N.S.; Gulley, J.L. Significance and implications of FDA approval of pembrolizumab for biomarker-defined disease. J. Immunother. Cancer 2018, 6, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Lesokhin, A.M.; Ansell, S.M.; Armand, P.; Scott, E.C.; Halwani, A.; Gutierrez, M.; Millenson, M.M.; Cohen, A.D.; Schuster, S.J.; Lebovic, D.; et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J. Clin. Oncol. 2016, 34, 2698–2704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  308. Armand, P.; Lesokhin, A.; Borrello, I.; Timmerman, J.; Gutierrez, M.; Zhu, L.; Popa McKiver, M.; Ansell, S.M. A phase 1b study of dual PD-1 and CTLA-4 or KIR blockade in patients with relapsed/refractory lymphoid malignancies. Leukemia 2021, 35, 777–786. [Google Scholar] [CrossRef] [PubMed]
  309. Bertamini, L.; Gay, F. Checkpoint inhibitors and myeloma: Promises, deadlocks and new directions. Ann. Transl. Med. 2020, 8, 777. [Google Scholar] [CrossRef]
  310. Casulo, C.; Santoro, A.; Ando, K.; Le Gouill, S.; Ruan, J.; Radford, J.; Arcaini, L.; Pinto, A.; Bouabdallah, R.; Izutsu, K.; et al. Durvalumab (Anti PD-L1) As Monotherapy or in Combination Therapy for Relapsed/Refractory (r/r) Diffuse Large B-Cell Lymphoma (DLBCL) and Follicular Lymphoma (FL): A Subgroup Analysis from the Phase 1/2 Fusion NHL-001 Global Multicenter Trial. Blood 2019, 134, 5320. [Google Scholar] [CrossRef]
  311. Ribrag, V.; Lee, S.T.; Rizzieri, D.; Dyer, M.J.S.; Fayad, L.; Kurzrock, R.; Andritsos, L.; Bouabdallah, R.; Hayat, A.; Bacon, L.; et al. A Phase 1b Study to Evaluate the Safety and Efficacy of Durvalumab in Combination with Tremelimumab or Danvatirsen in Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma. Clin. Lymphoma Myeloma Leuk. 2021, 21, 309–317.e3. [Google Scholar] [CrossRef]
  312. Hirayama, A.V.; Gauthier, J.; Hay, K.A.; Sheih, A.; Cherian, S.; Chen, X.; Pender, B.S.; Hawkins, R.M.; Vakil, A.; Steinmetz, R.N.; et al. Efficacy and Toxicity of JCAR014 in Combination with Durvalumab for the Treatment of Patients with Relapsed/Refractory Aggressive B-Cell Non-Hodgkin Lymphoma. Blood 2018, 132, 1680. [Google Scholar] [CrossRef]
  313. Kaiser, L.M.; Hunter, Z.R.; Treon, S.P.; Buske, C. CXCR4 in Waldenström’s Macroglobulinema: Chances and challenges. Leukemia 2021, 35, 333–345. [Google Scholar] [CrossRef] [PubMed]
  314. Kashyap, M.K.; Kumar, D.; Jones, H.; Amaya-Chanaga, C.I.; Choi, M.Y.; Melo-Cardenas, J.; Ale-Ali, A.; Kuhne, M.R.; Sabbatini, P.; Cohen, L.J.; et al. Ulocuplumab (BMS-936564/MDX1338): A fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget 2016, 7, 2809–2822. [Google Scholar] [CrossRef] [Green Version]
  315. Roccaro, A.M.; Sacco, A.; Jimenez, C.; Maiso, P.; Moschetta, M.; Mishima, Y.; Aljawai, Y.; Sahin, I.; Kuhne, M.; Cardarelli, P.; et al. C1013G/CXCR4 acts as a driver mutation of tumor progression and modulator of drug resistance in lymphoplasmacytic lymphoma. Blood 2014, 123, 4120–4131. [Google Scholar] [CrossRef]
  316. Ghobrial, I.M.; Liu, C.J.; Redd, R.A.; Perez, R.P.; Baz, R.; Zavidij, O.; Sklavenitis-Pistofidis, R.; Richardson, P.G.; Anderson, K.C.; Laubach, J.; et al. A Phase Ib/II Trial of the First-in-Class Anti-CXCR4 Antibody Ulocuplumab in Combination with Lenalidomide or Bortezomib Plus Dexamethasone in Relapsed Multiple Myeloma. Clin. Cancer Res. 2020, 26, 344–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  317. Rasche, L.; Wäsch, R.; Munder, M.; Goldschmidt, H.; Raab, M.S. Novel immunotherapies in multiple myeloma—Chances and challenges. Haematologica 2021, 106, 2555–2565. [Google Scholar] [CrossRef] [PubMed]
  318. Gormley, N.J.; Ko, C.W.; Deisseroth, A.; Nie, L.; Kaminskas, E.; Kormanik, N.; Goldberg, K.B.; Farrell, A.T.; Pazdur, R. FDA Drug Approval: Elotuzumab in Combination with Lenalidomide and Dexamethasone for the Treatment of Relapsed or Refractory Multiple Myeloma. Clin. Cancer Res. 2017, 23, 6759–6763. [Google Scholar] [CrossRef] [Green Version]
  319. Bauvois, B. Transmembrane proteases in focus: Diversity and redundancy? J. Leukoc. Biol. 2001, 70, 11–17. [Google Scholar] [CrossRef]
  320. Verhulst, E.; Garnier, D.; De Meester, I.; Bauvois, B. Validating Cell Surface Proteases as Drug Targets for Cancer Therapy: What Do We Know, and Where Do We Go? Cancers 2022, 14, 624. [Google Scholar] [CrossRef] [PubMed]
  321. Schliemann, C.; Gerwing, M.; Heinzow, H.; Harrach, S.; Schwöppe, C.; Wildgruber, M.; Hansmeier, A.A.; Angenendt, L.; Berdel, A.F.; Stalmann, U.; et al. First-In-Class CD13-Targeted Tissue Factor tTF-NGR in Patients with Recurrent or Refractory Malignant Tumors: Results of a Phase I Dose-Escalation Study. Cancers 2020, 12, 1488. [Google Scholar] [CrossRef] [PubMed]
  322. Berglund, Å.; Ullén, A.; Lisyanskaya, A.; Orlov, S.; Hagberg, H.; Tholander, B.; Lewensohn, R.; Nygren, P.; Spira, J.; Harmenberg, J.; et al. First-in-human, phase I/IIa clinical study of the peptidase potentiated alkylator melflufen administered every three weeks to patients with advanced solid tumor malignancies. Investig. New Drugs 2015, 33, 1232–1241. [Google Scholar] [CrossRef]
  323. Richardson, P.G.; Oriol, A.; Larocca, A.; Bladé, J.; Cavo, M.; Rodriguez-Otero, P.; Leleu, X.; Nadeem, O.; Hiemenz, J.W.; Hassoun, H.; et al. Melflufen and Dexamethasone in Heavily Pretreated Relapsed and Refractory Multiple Myeloma. J. Clin. Oncol. 2021, 39, 757–767. [Google Scholar] [CrossRef] [PubMed]
  324. Kudo, K.; Imai, C.; Lorenzini, P.; Kamiya, T.; Kono, K.; Davidoff, A.M.; Chng, W.J.; Campana, D. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 2014, 74, 93–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Munoz, J.; Jaglowski, S.; McKinney, M.S.; Isufi, I.; Stiff, P.J.; Sachs, J.; Ranger, A.; Harris, P.; Payumo, F.; Akard, L.P. A Phase 1 Study of ACTR087 in Combination with Rituximab, in Subjects with Relapsed or Refractory CD20-Positive B-Cell Lymphoma. Blood 2019, 134, 244. [Google Scholar] [CrossRef]
  326. Merli, M.; Ferrario, A.; Maffioli, M.; Olivares, C.; Stasia, A.; Arcaini, L.; Passamonti, F. New uses for brentuximab vedotin and novel antibody drug conjugates in lymphoma. Expert Rev. Hematol. 2016, 9, 767–780. [Google Scholar] [CrossRef]
  327. Grover, N.S.; Savoldo, B. Challenges of driving CD30-directed CAR-T cells to the clinic. BMC Cancer 2019, 19, 203. [Google Scholar] [CrossRef] [Green Version]
  328. Rodrigues-Fernandes, C.I.; Abreu, L.G.; Radhakrishnan, R.; Perez, D.; Amaral-Silva, G.K.; Gondak, R.O.; Rahimi, S.; Brennan, P.A.; Fonseca, F.P.; Vargas, P.A. Prognostic significance of CD30 expression in diffuse large B-cell lymphoma: A systematic review with meta-analysis. J. Oral Pathol. Med. 2021, 50, 587–593. [Google Scholar] [CrossRef] [PubMed]
  329. Atrash, S.; Moyo, T.K. A Review of Chimeric Antigen Receptor T-Cell Therapy for Myeloma and Lymphoma. OncoTargets Ther. 2021, 14, 2185–2201. [Google Scholar] [CrossRef] [PubMed]
  330. Ho, C.; Ruella, M.; Levine, B.L.; Svoboda, J. Adoptive T-cell therapy for Hodgkin lymphoma. Blood Adv. 2021, 5, 4291–4302. [Google Scholar] [CrossRef]
  331. Witkowska, M.; Smolewski, P.; Robak, T. Investigational therapies targeting CD37 for the treatment of B-cell lymphoid malignancies. Expert Opin. Investig. Drugs 2018, 27, 171–177. [Google Scholar] [CrossRef] [PubMed]
  332. Payandeh, Z.; Noori, E.; Khalesi, B.; Mard-Soltani, M.; Abdolalizadeh, J.; Khalili, S. Anti-CD37 targeted immunotherapy of B-Cell malignancies. Biotechnol. Lett. 2018, 40, 1459–1466. [Google Scholar] [CrossRef] [PubMed]
  333. Bobrowicz, M.; Kubacz, M.; Slusarczyk, A.; Winiarska, M. CD37 in B Cell Derived Tumors-More than Just a Docking Point for Monoclonal Antibodies. Int. J. Mol. Sci. 2020, 21, 9531. [Google Scholar] [CrossRef] [PubMed]
  334. Danilov, A.V.; Spurgeon, S.E.; Siddiqi, T.; Quinson, A.M.; Maier, D.; Smith, D.; Brown, J.R. A phase Ib, open label, dose escalation trial of the anti-CD37 monoclonal antibody, BI 836826, in combination with ibrutinib in patients with relapsed/refractory chronic lymphocytic leukemia. Investig. New Drugs 2021, 39, 1099–1105. [Google Scholar] [CrossRef] [PubMed]
  335. Yin, Z.; Zhang, Y.; Wang, X. Advances in chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Biomark Res. 2021, 9, 58. [Google Scholar] [CrossRef]
  336. Kroschinsky, F.; Middeke, J.M.; Janz, M.; Lenz, G.; Witzens-Harig, M.; Bouabdallah, R.; La Rosée, P.; Viardot, A.; Salles, G.; Kim, S.J.; et al. Phase I dose escalation study of BI 836826 (CD37 antibody) in patients with relapsed or refractory B-cell non-Hodgkin lymphoma. Investig. New Drugs 2020, 38, 1472–1482. [Google Scholar] [CrossRef] [PubMed]
  337. Stathis, A.; Flinn, I.W.; Madan, S.; Maddocks, K.; Freedman, A.; Weitman, S.; Zucca, E.; Munteanu, M.C.; Lia Palomba, M. Safety, tolerability, and preliminary activity of IMGN529, a CD37-targeted antibody-drug conjugate, in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: A dose-escalation, phase I study. Investig. New Drugs 2018, 36, 869–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  338. Levy, M.Y.; Jagadeesh, D.; Grudeva-Popova, Z.; Trněný, M.; Jurczak, W.; Pylypenko, H.; André, M.; Nasta, S.D.; Rechavi-Robinson, D.; Toffanin, S.; et al. Safety and Efficacy of CD37-Targeting Naratuximab Emtansine PLUS Rituximab in Diffuse Large B-Cell Lymphoma and Other NON-Hodgkin’S B-Cell Lymphomas-a Phase 2 Study. Blood 2021, 138, 526. [Google Scholar] [CrossRef]
  339. Sawas, A.; Savage, K.J.; Perez, R.P.; Advani, R.H.; Melhem-Bertrandt, A.; Lackey, J.; Trave, F.; Anand, B.; Huang, Y.; Vincent, M.; et al. A first in human experience of the anti-CD37 antibody-drug conjugate AGS67E in lymphoid malignancies. J. Clin. Oncol. 2016, 34, 7549. [Google Scholar] [CrossRef]
  340. Elvington, M.; Liszewski, M.K.; Atkinson, J.P. CD46 and Oncologic Interactions: Friendly Fire against Cancer. Antibodies 2020, 9, 59. [Google Scholar] [CrossRef] [PubMed]
  341. Wong, S.; Imus, P.; Mark, T.; Kaufman, J.; Imus, A.; Zonder, J.A.; Walker, Z.; Sherbenou, D.; Schroeder, M.; Abbey, J.; et al. P-225: A first-in-human study of FOR46 in patients with triple refractory Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2021, 21, 164. [Google Scholar] [CrossRef]
  342. Sick, E.; Jeanne, A.; Schneider, C.; Dedieu, S.; Takeda, K.; Martiny, L. CD47 update: A multifaceted actor in the tumour microenvironment of potential therapeutic interest. Br. J. Pharmacol. 2012, 167, 1415–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  343. Maute, R.; Xu, J.; Weissman, I.L. CD47-SIRPα-targeted therapeutics: Status and prospects. Immuno. Oncol. Technol. 2022, 13, 100070. [Google Scholar] [CrossRef]
  344. Jiang, Z.; Sun, H.; Yu, J.; Tian, W.; Song, Y. Targeting CD47 for cancer immunotherapy. J. Hematol. Oncol. 2021, 14, 180. [Google Scholar] [CrossRef]
  345. Saygin, C.; Carraway, H.E. Current and emerging strategies for management of myelodysplastic syndromes. Blood Rev. 2021, 48, 100791. [Google Scholar] [CrossRef]
  346. Gilead. Safety Concerns Prompt Pause of Magrolimab Trials. Cancer Discov. 2022, 12, 877–878. [Google Scholar] [CrossRef] [PubMed]
  347. Lambert, J.M.; O’Leary, J.; Whiteman, K.R.; Goldmacher, V.S. Targeting CD56 (NCAM)-Expressing Neoplasms with Lorvotuzumab Mertansine. In Antibody-Drug Conjugates and Immunotoxins; Springer: New York, NY, USA, 2012; pp. 273–293. [Google Scholar]
  348. Ailawadhi, S.; Kelly, K.R.; Vescio, R.A.; Jagannath, S.; Wolf, J.; Gharibo, M.; Sher, T.; Bojanini, L.; Kirby, M.; Chanan-Khan, A. A Phase I Study to Assess the Safety and Pharmacokinetics of Single-agent Lorvotuzumab Mertansine (IMGN901) in Patients with Relapsed and/or Refractory CD-56-positive Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2019, 19, 29–34. [Google Scholar] [CrossRef]
  349. Gil-Yarom, N.; Radomir, L.; Sever, L.; Kramer, M.P.; Lewinsky, H.; Bornstein, C.; Blecher-Gonen, R.; Barnett-Itzhaki, Z.; Mirkin, V.; Friedlander, G.; et al. CD74 is a novel transcription regulator. Proc. Natl. Acad. Sci. USA 2017, 114, 562–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  350. Stein, R.; Mattes, M.J.; Cardillo, T.M.; Hansen, H.J.; Chang, C.H.; Burton, J.; Govindan, S.; Goldenberg, D.M. CD74: A new candidate target for the immunotherapy of B-cell neoplasms. Clin. Cancer Res. 2007, 13, 5556s–5563s. [Google Scholar] [CrossRef] [PubMed]
  351. Kaufman, J.L.; Niesvizky, R.; Stadtmauer, E.A.; Chanan-Khan, A.; Siegel, D.; Horne, H.; Wegener, W.A.; Goldenberg, D.M. Phase I, multicentre, dose-escalation trial of monotherapy with milatuzumab (humanized anti-CD74 monoclonal antibody) in relapsed or refractory multiple myeloma. Br. J. Haematol. 2013, 163, 478–486. [Google Scholar] [CrossRef] [PubMed]
  352. Abrahams, C.L.; Li, X.; Embry, M.; Yu, A.; Krimm, S.; Krueger, S.; Greenland, N.Y.; Wen, K.W.; Jones, C.; DeAlmeida, V.; et al. Targeting CD74 in multiple myeloma with the novel, site-specific antibody-drug conjugate STRO-001. Oncotarget 2018, 9, 37700–37714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. Sawalha, Y.; Maddocks, K. Profile of Polatuzumab Vedotin in the Treatment of Patients with Relapsed/Refractory Non-Hodgkin Lymphoma: A Brief Report on the Emerging Clinical Data. Onco Targets Ther. 2020, 13, 5123–5133. [Google Scholar] [CrossRef] [PubMed]
  354. Arribas, J.; Esselens, C. ADAM17 as a therapeutic target in multiple diseases. Curr. Pharm. Des. 2009, 15, 2319–2335. [Google Scholar] [CrossRef]
  355. Witters, L.; Scherle, P.; Friedman, S.; Fridman, J.; Caulder, E.; Newton, R.; Lipton, A. Synergistic inhibition with a dual epidermal growth factor receptor/HER-2/neu tyrosine kinase inhibitor and a disintegrin and metalloprotease inhibitor. Cancer Res. 2008, 68, 7083–7089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  356. Bachanova, V.; Kolla, B.; Cao, Q.; Weisdorf, D.; Rashidi, A.; Warlick, E.; El Jurdi, N.; Wangen, R.; Arora, M.; Brunstein, C.; et al. ADAM17 Inhibitor INCB7839 With Rituximab As Consolidation After Autologous Hct For Diffuse Large B Cell Lymphoma: A Novel Relapse Prevention Strategy. In Proceedings of the Virtual 46th Annual Meeting of the EBMT, Virtual, 29 August–1 September 2020; p. 133. Available online: https://www.ebmt.org/46th-annual-meeting-ebmt (accessed on 6 November 2022).
  357. Polson, A.G.; Zheng, B.; Elkins, K.; Chang, W.; Du, C.; Dowd, P.; Yen, L.; Tan, C.; Hongo, J.A.; Koeppen, H.; et al. Expression pattern of the human FcRH/IRTA receptors in normal tissue and in B-chronic lymphocytic leukemia. Int. Immunol. 2006, 18, 1363–1373. [Google Scholar] [CrossRef] [Green Version]
  358. Ise, T.; Nagata, S.; Kreitman, R.J.; Wilson, W.H.; Wayne, A.S.; Stetler-Stevenson, M.; Bishop, M.R.; Scheinberg, D.A.; Rassenti, L.; Kipps, T.J.; et al. Elevation of soluble CD307 (IRTA2/FcRH5) protein in the blood and expression on malignant cells of patients with multiple myeloma, chronic lymphocytic leukemia, and mantle cell lymphoma. Leukemia 2007, 21, 169–174. [Google Scholar] [CrossRef]
  359. Smith, E.L.; Harrington, K.; Staehr, M.; Masakayan, R.; Jones, J.; Long, T.J.; Ng, K.Y.; Ghoddusi, M.; Purdon, T.J.; Wang, X.; et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci. Transl. Med. 2019, 11, eaau7746. [Google Scholar] [CrossRef] [PubMed]
  360. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  361. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  362. Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [Green Version]
  363. Kirsch, B.J.; Chang, S.J.; Betenbaugh, M.J.; Le, A. Non-Hodgkin Lymphoma Metabolism. Adv. Exp. Med. Biol. 2021, 1311, 103–116. [Google Scholar]
  364. Bhalla, K.; Jaber, S.; Nahid, M.N.; Underwood, K.; Beheshti, A.; Landon, A.; Bhandary, B.; Bastian, P.; Evens, A.M.; Haley, J.; et al. Role of hypoxia in Diffuse Large B-cell Lymphoma: Metabolic repression and selective translation of HK2 facilitates development of DLBCL. Sci. Rep. 2018, 8, 744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  365. Nakajima, K.; Kawashima, I.; Koshiisi, M.; Kumagai, T.; Suzuki, M.; Suzuki, J.; Mitsumori, T.; Kirito, K. Glycolytic enzyme hexokinase II is a putative therapeutic target in B-cell malignant lymphoma. Exp. Hematol. 2019, 78, 46–55.e3. [Google Scholar] [CrossRef]
  366. Lu, J.; Böttcher, M.; Walther, T.; Mougiakakos, D.; Zenz, T.; Huber, W. Energy metabolism is co-determined by genetic variants in chronic lymphocytic leukemia and influences drug sensitivity. Haematologica 2019, 104, 1830–1840. [Google Scholar] [CrossRef] [PubMed]
  367. Adekola, K.U.; Dalva Aydemir, S.; Ma, S.; Zhou, Z.; Rosen, S.T.; Shanmugam, M. Investigating and targeting chronic lymphocytic leukemia metabolism with the human immunodeficiency virus protease inhibitor ritonavir and metformin. Leuk. Lymphoma 2015, 56, 450–459. [Google Scholar] [CrossRef] [Green Version]
  368. Dalva-Aydemir, S.; Bajpai, R.; Martinez, M.; Adekola, K.U.; Kandela, I.; Wei, C.; Singhal, S.; Koblinski, J.E.; Raje, N.S.; Rosen, S.T.; et al. Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin. Clin. Cancer Res. 2015, 21, 1161–1171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  369. Zhao, H.; Swanson, K.D.; Zheng, B. Therapeutic Repurposing of Biguanides in Cancer. Trends Cancer 2021, 7, 714–730. [Google Scholar] [CrossRef]
  370. Hartmann, S.; Agostinelli, C.; Diener, J.; Döring, C.; Fanti, S.; Zinzani, P.L.; Gallamini, A.; Bergmann, L.; Pileri, S.; Hansmann, M.L. GLUT1 expression patterns in different Hodgkin lymphoma subtypes and progressively transformed germinal centers. BMC Cancer 2012, 12, 586. [Google Scholar] [CrossRef] [Green Version]
  371. Afonso, J.; Pinto, T.; Simões-Sousa, S.; Schmitt, F.; Longatto-Filho, A.; Pinheiro, C.; Marques, H.; Baltazar, F. Clinical significance of metabolism-related biomarkers in non-Hodgkin lymphoma-MCT1 as potential target in diffuse large B cell lymphoma. Cell Oncol. (Dordr.) 2019, 42, 303–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  372. Noble, R.A.; Bell, N.; Blair, H.; Sikka, A.; Thomas, H.; Phillips, N.; Nakjang, S.; Miwa, S.; Crossland, R.; Rand, V.; et al. Inhibition of monocarboxyate transporter 1 by AZD3965 as a novel therapeutic approach for diffuse large B-cell lymphoma and Burkitt lymphoma. Haematologica 2017, 102, 1247–1257. [Google Scholar] [CrossRef]
  373. Halford, S.E.; Walter, H.; McKay, P.; Townsend, W.; Linton, K.; Heinzmann, K.; Dragoni, I.; Brotherton, L.; Veal, G.; Siskos, A.; et al. Phase I expansion study of the first-in-class monocarboxylate transporter 1 (MCT1) inhibitor AZD3965 in patients with diffuse large B-cell lymphoma (DLBCL) and Burkitt lymphoma (BL). J. Clin. Oncol. 2021, 39, 3115. [Google Scholar] [CrossRef]
  374. Xiao, G.; Chan, L.N.; Klemm, L.; Braas, D.; Chen, Z.; Geng, H.; Zhang, Q.C.; Aghajanirefah, A.; Cosgun, K.N.; Sadras, T.; et al. B-Cell-Specific Diversion of Glucose Carbon Utilization Reveals a Unique Vulnerability in B Cell Malignancies. Cell 2018, 173, 470–484.e18. [Google Scholar] [CrossRef] [PubMed]
  375. Zhang, L.; Yao, Y.; Zhang, S.; Liu, Y.; Guo, H.; Ahmed, M.; Bell, T.; Zhang, H.; Han, G.; Lorence, E.; et al. Metabolic reprogramming toward oxidative phosphorylation identifies a therapeutic target for mantle cell lymphoma. Sci. Transl. Med. 2019, 11, eaau1167. [Google Scholar] [CrossRef]
  376. Zhan, X.; Yu, W.; Franqui-Machin, R.; Bates, M.L.; Nadiminti, K.; Cao, H.; Amendt, B.A.; Jethava, Y.; Frech, I.; Zhan, F.; et al. Alteration of mitochondrial biogenesis promotes disease progression in multiple myeloma. Oncotarget 2017, 8, 111213–111224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  377. Galicia-Vázquez, G.; Aloyz, R. Metabolic rewiring beyond Warburg in chronic lymphocytic leukemia: How much do we actually know? Crit. Rev Oncol. Hematol. 2019, 134, 65–70. [Google Scholar] [CrossRef] [PubMed]
  378. Nie, Y.; Yun, X.; Zhang, Y.; Wang, X. Targeting metabolic reprogramming in chronic lymphocytic leukemia. Exp. Hematol. Oncol. 2022, 11, 39. [Google Scholar] [CrossRef] [PubMed]
  379. Jitschin, R.; Hofmann, A.D.; Bruns, H.; Gießl, A.; Bricks, J.; Berger, J.; Saul, D.; Eckart, M.J.; Mackensen, A.; Mougiakakos, D. Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic targets in chronic lymphocytic leukemia. Blood 2014, 123, 2663–2672. [Google Scholar] [CrossRef] [Green Version]
  380. Ecker, V.; Stumpf, M.; Brandmeier, L.; Neumayer, T.; Pfeuffer, L.; Engleitner, T.; Ringshausen, I.; Nelson, N.; Jücker, M.; Wanninger, S.; et al. Targeted PI3K/AKT-hyperactivation induces cell death in chronic lymphocytic leukemia. Nat. Commun. 2021, 12, 3526. [Google Scholar] [CrossRef]
  381. Noble, R.A.; Thomas, H.; Zhao, Y.; Herendi, L.; Howarth, R.; Dragoni, I.; Keun, H.C.; Vellano, C.P.; Marszalek, J.R.; Wedge, S.R. Simultaneous targeting of glycolysis and oxidative phosphorylation as a therapeutic strategy to treat diffuse large B-cell lymphoma. Br. J. Cancer 2022, 127, 937–947. [Google Scholar] [CrossRef]
  382. Zachar, Z.; Marecek, J.; Maturo, C.; Gupta, S.; Stuart, S.D.; Howell, K.; Schauble, A.; Lem, J.; Piramzadian, A.; Karnik, S.; et al. Non-redox-active lipoate derivates disrupt cancer cell mitochondrial metabolism and are potent anticancer agents in vivo. J. Mol. Med. 2011, 89, 1137–1148. [Google Scholar] [CrossRef]
  383. Stuart, S.D.; Schauble, A.; Gupta, S.; Kennedy, A.D.; Keppler, B.R.; Bingham, P.M.; Zachar, Z. A strategically designed small molecule attacks alpha-ketoglutarate dehydrogenase in tumor cells through a redox process. Cancer Metab. 2014, 2, 4. [Google Scholar] [CrossRef] [Green Version]
  384. Ambrosio, M.R.; Piccaluga, P.P.; Ponzoni, M.; Rocca, B.J.; Malagnino, V.; Onorati, M.; De Falco, G.; Calbi, V.; Ogwang, M.; Naresh, K.N.; et al. The alteration of lipid metabolism in Burkitt lymphoma identifies a novel marker: Adipophilin. PLoS ONE 2012, 7, e44315. [Google Scholar] [CrossRef] [PubMed]
  385. Heintel, D.; Kienle, D.; Shehata, M.; Kröber, A.; Kroemer, E.; Schwarzinger, I.; Mitteregger, D.; Le, T.; Gleiss, A.; Mannhalter, C.; et al. High expression of lipoprotein lipase in poor risk B-cell chronic lymphocytic leukemia. Leukemia 2005, 19, 1216–1223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  386. Rozovski, U.; Grgurevic, S.; Bueso-Ramos, C.; Harris, D.M.; Li, P.; Liu, Z.; Wu, J.Y.; Jain, P.; Wierda, W.; Burger, J.; et al. Aberrant LPL expression, driven by STAT3, Mediates free fatty acid metabolism in cll cells. Mol. Cancer Res. 2015, 13, 944–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  387. Pallasch, C.P.; Schwamb, J.; Königs, S.; Schulz, A.; Debey, S.; Kofler, D.; Schultze, J.L.; Hallek, M.; Ultsch, A.; Wendtner, C.M. Targeting lipid metabolism by the lipoprotein lipase inhibitor orlistat results in apoptosis of B-cell chronic lymphocytic leukemia cells. Leukemia 2008, 22, 585–592. [Google Scholar] [CrossRef] [Green Version]
  388. Gelebart, P.; Zak, Z.; Anand, M.; Belch, A.; Lai, R. Blockade of fatty acid synthase triggers significant apoptosis in mantle cell lymphoma. PLoS ONE 2012, 7, e33738. [Google Scholar] [CrossRef] [Green Version]
  389. Uddin, S.; Hussain, A.R.; Ahmed, M.; Abubaker, J.; Al-Sanea, N.; Abduljabbar, A.; Ashari, L.H.; Alhomoud, S.; Al-Dayel, F.; Bavi, P.; et al. High prevalence of fatty acid synthase expression in colorectal cancers in Middle Eastern patients and its potential role as a therapeutic target. Am. J. Gastroenterol. 2009, 104, 1790–1801. [Google Scholar] [CrossRef]
  390. Rozovski, U.; Harris, D.M.; Li, P.; Liu, Z.; Jain, P.; Ferrajoli, A.; Burger, J.; Thompson, P.; Jain, N.; Wierda, W.; et al. STAT3-activated CD36 facilitates fatty acid uptake in chronic lymphocytic leukemia cells. Oncotarget 2018, 9, 21268–21280. [Google Scholar] [CrossRef]
  391. Liu, P.P.; Liu, J.; Jiang, W.Q.; Carew, J.S.; Ogasawara, M.A.; Pelicano, H.; Croce, C.M.; Estrov, Z.; Xu, R.H.; Keating, M.J.; et al. Elimination of chronic lymphocytic leukemia cells in stromal microenvironment by targeting CPT with an antiangina drug perhexiline. Oncogene 2016, 35, 5663–5673. [Google Scholar] [CrossRef]
  392. Bolzoni, M.; Chiu, M.; Accardi, F.; Vescovini, R.; Airoldi, I.; Storti, P.; Todoerti, K.; Agnelli, L.; Missale, G.; Andreoli, R.; et al. Dependence on glutamine uptake and glutamine addiction characterize myeloma cells: A new attractive target. Blood 2016, 128, 667–679. [Google Scholar] [CrossRef]
  393. Galicia-Vázquez, G.; Smith, S.; Aloyz, R. Del11q-positive CLL lymphocytes exhibit altered glutamine metabolism and differential response to GLS1 and glucose metabolism inhibition. Blood Cancer J. 2018, 8, 13. [Google Scholar] [CrossRef] [Green Version]
  394. Le, A.; Lane, A.N.; Hamaker, M.; Bose, S.; Gouw, A.; Barbi, J.; Tsukamoto, T.; Rojas, C.J.; Slusher, B.S.; Zhang, H.; et al. Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. 2012, 15, 110–121. [Google Scholar] [CrossRef] [Green Version]
  395. Anastasiou, D. Tumour microenvironment factors shaping the cancer metabolism landscape. Br. J. Cancer 2017, 116, 277–286. [Google Scholar] [CrossRef] [PubMed]
  396. Liu, Y.; Zhou, X.; Wang, X. Targeting the tumor microenvironment in B-cell lymphoma: Challenges and opportunities. J. Hematol. Oncol. 2021, 14, 125. [Google Scholar] [CrossRef] [PubMed]
  397. Vangapandu, H.V.; Ayres, M.L.; Bristow, C.A.; Wierda, W.G.; Keating, M.J.; Balakrishnan, K.; Stellrecht, C.M.; Gandhi, V. The Stromal Microenvironment Modulates Mitochondrial Oxidative Phosphorylation in Chronic Lymphocytic Leukemia Cells. Neoplasia 2017, 19, 762–771. [Google Scholar] [CrossRef] [PubMed]
  398. Yosifov, D.Y.; Idler, I.; Bhattacharya, N.; Reichenzeller, M.; Close, V.; Ezerina, D.; Scheffold, A.; Jebaraj, B.M.C.; Kugler, S.; Bloehdorn, J.; et al. Oxidative stress as candidate therapeutic target to overcome microenvironmental protection of CLL. Leukemia 2020, 34, 115–127. [Google Scholar] [CrossRef] [PubMed]
  399. Zhang, W.; Trachootham, D.; Liu, J.; Chen, G.; Pelicano, H.; Garcia-Prieto, C.; Lu, W.; Burger, J.A.; Croce, C.M.; Plunkett, W.; et al. Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat. Cell Biol. 2012, 14, 276–286. [Google Scholar] [CrossRef] [Green Version]
  400. Chen, Z.; Simon-Molas, H.; Cretenet, G.; Valle-Argos, B.; Smith, L.D.; Forconi, F.; Schomakers, B.V.; van Weeghel, M.; Bryant, D.J.; van Bruggen, J.A.C.; et al. Characterization of metabolic alterations of chronic lymphocytic leukemia in the lymph node microenvironment. Blood 2022, 140, 630–643. [Google Scholar] [CrossRef]
  401. Burt, R.; Dey, A.; Aref, S.; Aguiar, M.; Akarca, A.; Bailey, K.; Day, W.; Hooper, S.; Kirkwood, A.; Kirschner, K.; et al. Activated stromal cells transfer mitochondria to rescue acute lymphoblastic leukemia cells from oxidative stress. Blood 2019, 134, 1415–1429. [Google Scholar] [CrossRef]
  402. Marlein, C.R.; Piddock, R.E.; Mistry, J.J.; Zaitseva, L.; Hellmich, C.; Horton, R.H.; Zhou, Z.; Auger, M.J.; Bowles, K.M.; Rushworth, S.A. CD38-Driven Mitochondrial Trafficking Promotes Bioenergetic Plasticity in Multiple Myeloma. Cancer Res. 2019, 79, 2285–2297. [Google Scholar] [CrossRef]
  403. Panaroni, C.; Fulzele, K.; Mori, T.; Siu, K.T.; Onyewadume, C.; Maebius, A.; Raje, N. Multiple myeloma cells induce lipolysis in adipocytes and uptake fatty acids through fatty acid transporter proteins. Blood 2022, 139, 876–888. [Google Scholar] [CrossRef]
  404. Pavlides, S.; Tsirigos, A.; Vera, I.; Flomenberg, N.; Frank, P.G.; Casimiro, M.C.; Wang, C.; Fortina, P.; Addya, S.; Pestell, R.G.; et al. Loss of stromal caveolin-1 leads to oxidative stress, mimics hypoxia and drives inflammation in the tumor microenvironment, conferring the “reverse Warburg effect”: A transcriptional informatics analysis with validation. Cell Cycle 2010, 9, 2201–2219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  405. Sotgia, F.; Whitaker-Menezes, D.; Martinez-Outschoorn, U.E.; Flomenberg, N.; Birbe, R.C.; Witkiewicz, A.K.; Howell, A.; Philp, N.J.; Pestell, R.G.; Lisanti, M.P. Mitochondrial metabolism in cancer metastasis: Visualizing tumor cell mitochondria and the “reverse Warburg effect” in positive lymph node tissue. Cell Cycle 2012, 11, 1445–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  406. Gooptu, M.; Whitaker-Menezes, D.; Sprandio, J.; Domingo-Vidal, M.; Lin, Z.; Uppal, G.; Gong, J.; Fratamico, R.; Leiby, B.; Dulau-Florea, A.; et al. Mitochondrial and glycolytic metabolic compartmentalization in diffuse large B-cell lymphoma. Semin. Oncol. 2017, 44, 204–217. [Google Scholar] [PubMed]
  407. Sakamoto, A.; Kunou, S.; Shimada, K.; Tsunoda, M.; Aoki, T.; Iriyama, C.; Tomita, A.; Nakamura, S.; Hayakawa, F.; Kiyoi, H. Pyruvate secreted from patient-derived cancer-associated fibroblasts supports survival of primary lymphoma cells. Cancer Sci. 2019, 110, 269–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  408. Mikkilineni, L.; Whitaker-Menezes, D.; Domingo-Vidal, M.; Sprandio, J.; Avena, P.; Cotzia, P.; Dulau-Florea, A.; Gong, J.; Uppal, G.; Zhan, T.; et al. Hodgkin lymphoma: A complex metabolic ecosystem with glycolytic reprogramming of the tumor microenvironment. Semin. Oncol. 2017, 44, 218–225. [Google Scholar] [CrossRef] [PubMed]
  409. Alfaifi, A.; Bahashwan, S.; Alsaadi, M.; Malhan, H.; Aqeel, A.; Al-Kahiry, W.; Almehdar, H.; Qadri, I. Metabolic Biomarkers in B-Cell Lymphomas for Early Diagnosis and Prediction, as Well as Their Influence on Prognosis and Treatment. Diagnostics 2022, 12, 394. [Google Scholar] [CrossRef]
  410. Liu, X.; Wang, L.; Jiang, W.; Lu, W.; Yang, J.; Yang, W. B cell lymphoma with different metabolic characteristics show distinct sensitivities to metabolic inhibitors. J. Cancer 2018, 9, 1582–1591. [Google Scholar] [CrossRef]
  411. Barberini, L.; Noto, A.; Fattuoni, C.; Satta, G.; Zucca, M.; Cabras, M.G.; Mura, E.; Cocco, P. The Metabolomic Profile of Lymphoma Subtypes: A Pilot Study. Molecules 2019, 24, 2367. [Google Scholar] [CrossRef] [Green Version]
  412. Puchades-Carrasco, L.; Lecumberri, R.; Martínez-López, J.; Lahuerta, J.J.; Mateos, M.V.; Prósper, F.; San-Miguel, J.F.; Pineda-Lucena, A. Multiple myeloma patients have a specific serum metabolomic profile that changes after achieving complete remission. Clin. Cancer Res. 2013, 19, 4770–4779. [Google Scholar] [CrossRef]
  413. Bai, Y.; Zhang, H.; Sun, X.; Sun, C.; Ren, L. Biomarker identification and pathway analysis by serum metabolomics of childhood acute lymphoblastic leukemia. Clin. Chim. Acta 2014, 436, 207–216. [Google Scholar] [CrossRef]
  414. MacIntyre, D.A.; Jiménez, B.; Lewintre, E.J.; Martín, C.R.; Schäfer, H.; Ballesteros, C.G.; Mayans, J.R.; Spraul, M.; García-Conde, J.; Pineda-Lucena, A. Serum metabolome analysis by 1H-NMR reveals differences between chronic lymphocytic leukaemia molecular subgroups. Leukemia 2010, 24, 788–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  415. Monti, S.; Savage, K.J.; Kutok, J.L.; Feuerhake, F.; Kurtin, P.; Mihm, M.; Wu, B.; Pasqualucci, L.; Neuberg, D.; Aguiar, R.C.; et al. Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood 2005, 105, 1851–1861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  416. Caro, P.; Kishan, A.U.; Norberg, E.; Stanley, I.A.; Chapuy, B.; Ficarro, S.B.; Polak, K.; Tondera, D.; Gounarides, J.; Yin, H.; et al. Metabolic signatures uncover distinct targets in molecular subsets of diffuse large B cell lymphoma. Cancer Cell 2012, 22, 547–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  417. Chen, L.; Monti, S.; Juszczynski, P.; Daley, J.; Chen, W.; Witzig, T.E.; Habermann, T.M.; Kutok, J.L.; Shipp, M.A. SYK-dependent tonic B-cell receptor signaling is a rational treatment target in diffuse large B-cell lymphoma. Blood 2008, 111, 2230–2237. [Google Scholar] [CrossRef] [Green Version]
  418. Chiche, J.; Reverso-Meinietti, J.; Mouchotte, A.; Rubio-Patiño, C.; Mhaidly, R.; Villa, E.; Bossowski, J.P.; Proics, E.; Grima-Reyes, M.; Paquet, A.; et al. GAPDH Expression Predicts the Response to R-CHOP, the Tumor Metabolic Status, and the Response of DLBCL Patients to Metabolic Inhibitors. Cell Metab. 2019, 29, 1243–1257.e10. [Google Scholar] [CrossRef]
  419. Lemberg, K.M.; Gori, S.S.; Tsukamoto, T.; Rais, R.; Slusher, B.S. Clinical development of metabolic inhibitors for oncology. J. Clin. Investig. 2022, 132, e148550. [Google Scholar] [CrossRef]
  420. Stine, Z.E.; Schug, Z.T.; Salvino, J.M.; Dang, C.V. Targeting cancer metabolism in the era of precision oncology. Nat. Rev. Drug Discov. 2022, 21, 141–162. [Google Scholar] [CrossRef]
  421. Pi, M.; Kuang, H.; Yue, C.; Yang, Q.; Wu, A.; Li, Y.; Assaraf, Y.G.; Yang, D.H.; Wu, S. Targeting metabolism to overcome cancer drug resistance: A promising therapeutic strategy for diffuse large B cell lymphoma. Drug Resist. Updates 2022, 61, 100822. [Google Scholar] [CrossRef]
  422. Gdynia, G.; Robak, T.; Kopitz, J.; Heller, A.; Grekova, S.; Duglova, K.; Laukemper, G.; Heinzel-Gutenbrunner, M.; Gutenbrunner, C.; Roth, W.; et al. Distinct Activities of Glycolytic Enzymes Identify Chronic Lymphocytic Leukemia Patients with a more Aggressive Course and Resistance to Chemo-Immunotherapy. eBioMedicine 2018, 32, 125–133. [Google Scholar] [CrossRef] [PubMed]
  423. Guièze, R.; Liu, V.M.; Rosebrock, D.; Jourdain, A.A.; Hernández-Sánchez, M.; Martinez Zurita, A.; Sun, J.; Ten Hacken, E.; Baranowski, K.; Thompson, P.A.; et al. Mitochondrial Reprogramming Underlies Resistance to BCL-2 Inhibition in Lymphoid Malignancies. Cancer Cell 2019, 36, 369–384.e13. [Google Scholar] [CrossRef]
  424. Bajpai, R.; Sharma, A.; Achreja, A.; Edgar, C.L.; Wei, C.; Siddiqa, A.A.; Gupta, V.A.; Matulis, S.M.; McBrayer, S.K.; Mittal, A.; et al. Electron transport chain activity is a predictor and target for venetoclax sensitivity in multiple myeloma. Nat. Commun. 2020, 11, 1228. [Google Scholar] [CrossRef] [Green Version]
  425. Chukkapalli, V.; Gordon, L.I.; Venugopal, P.; Borgia, J.A.; Karmali, R. Metabolic changes associated with metformin potentiates Bcl-2 inhibitor, Venetoclax, and CDK9 inhibitor, BAY1143572 and reduces viability of lymphoma cells. Oncotarget 2018, 9, 21166–21181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  426. Ravera, S.; Ghiotto, F.; Tenca, C.; Gugiatti, E.; Santamaria, S.; Ledda, B.; Ibatici, A.; Cutrona, G.; Mazzarello, A.N.; Bagnara, D.; et al. Berberine affects mitochondrial activity and cell growth of leukemic cells from chronic lymphocytic leukemia patients. Sci. Rep. 2020, 10, 16519. [Google Scholar] [CrossRef]
  427. Vangapandu, H.V.; Havranek, O.; Ayres, M.L.; Kaipparettu, B.A.; Balakrishnan, K.; Wierda, G.D.W.; Keating, M.J.; Eric Davis, R.; Stellrecht, C.M.; Gandhi, V.; et al. B-cell receptor signaling regulates metabolism in chronic lymphocytic leukemia. Mol. Cancer Res. 2017, 15, 1692–1703. [Google Scholar] [CrossRef] [Green Version]
  428. Chowdhury, R.S.; Bouchard, E.D.J.; Saleh, R.; Nugent, Z.; Peltier, C.; Mejia, E.; Hou, S.; McFall, C.; Squires, M.; Hewitt, D.; et al. Mitochondrial Respiration Correlates with Prognostic Markers in Chronic Lymphocytic Leukemia and Is Normalized by Ibrutinib Treatment. Cancers 2020, 12, 650. [Google Scholar] [CrossRef] [Green Version]
  429. Lee, S.C.; Shestov, A.A.; Guo, L.; Zhang, Q.; Roman, J.C.; Liu, X.; Wang, H.Y.; Pickup, S.; Nath, K.; Lu, P.; et al. Metabolic Detection of Bruton’s Tyrosine Kinase Inhibition in Mantle Cell Lymphoma Cells. Mol. Cancer Res. 2019, 17, 1365–1377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  430. Yang, M. B cell lymphoma R-CHOPped by metabolic inhibitors. Sci. Transl. Med. 2019, 11, 117. [Google Scholar] [CrossRef]
  431. Gu, J.J.; Singh, A.; Xue, K.; Mavis, C.; Barth, M.; Yanamadala, V.; Lenz, P.; Grau, M.; Lenz, G.; Czuczman, M.S.; et al. Up-regulation of hexokinase II contributes to rituximab-chemotherapy resistance and is a clinically relevant target for therapeutic development. Oncotarget 2018, 9, 4020–4033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  432. Curtis, N.J.; Mooney, L.; Hopcroft, L.; Michopoulos, F.; Whalley, N.; Zhong, H.; Murray, C.; Logie, A.; Revill, M.; Byth, K.F.; et al. Pre-clinical pharmacology of AZD3965, a selective inhibitor of MCT1: DLBCL, NHL and Burkitt’s lymphoma anti-tumor activity. Oncotarget 2017, 8, 69219–69236. [Google Scholar] [CrossRef] [PubMed]
  433. Singh, A.R.; Gu, J.J.; Zhang, Q.; Torka, P.; Sundaram, S.; Mavis, C.; Hernandez-Ilizaliturri, F.J. Metformin sensitizes therapeutic agents and improves outcome in pre-clinical and clinical diffuse large B-cell lymphoma. Cancer Metab. 2020, 8, 10. [Google Scholar] [CrossRef] [PubMed]
  434. Jiang, D.; Mo, Q.; Sun, X.; Wang, X.; Dong, M.; Zhang, G.; Chen, F.; Zhao, Q. Pyruvate dehydrogenase kinase 4-mediated metabolic reprogramming is involved in rituximab resistance in diffuse large B-cell lymphoma by affecting the expression of MS4A1/CD20. Cancer Sci. 2021, 112, 3585–3597. [Google Scholar] [CrossRef] [PubMed]
  435. Ricci, J.E.; Chiche, J. Metabolic Reprogramming of Non-Hodgkin’s B-Cell Lymphomas and Potential Therapeutic Strategies. Front. Oncol. 2018, 8, 556. [Google Scholar] [CrossRef] [PubMed]
  436. Thompson, R.M.; Dytfeld, D.; Reyes, L.; Robinson, R.M.; Smith, B.; Manevich, Y.; Jakubowiak, A.; Komarnicki, M.; Przybylowicz-Chalecka, A.; Szczepaniak, T.; et al. Glutaminase inhibitor CB-839 synergizes with carfilzomib in resistant multiple myeloma cells. Oncotarget 2017, 8, 35863. [Google Scholar] [CrossRef] [Green Version]
  437. Sanchez, W.Y.; McGee, S.L.; Connor, T.; Mottram, B.; Wilkinson, A.; Whitehead, J.P.; Vuckovic, S.; Catley, L. Dichloroacetate inhibits aerobic glycolysis in multiple myeloma cells and increases sensitivity to bortezomib. Br. J. Cancer 2013, 108, 1624–1633. [Google Scholar] [CrossRef] [PubMed]
  438. Böttcher, M.; Baur, R.; Stoll, A.; Mackensen, A.; Mougiakakos, D. Linking Immunoevasion and Metabolic Reprogramming in B-Cell-Derived Lymphomas. Front. Oncol. 2020, 10, 594782. [Google Scholar] [CrossRef]
  439. Beielstein, A.C.; Pallasch, C.P. Tumor Metabolism as a Regulator of Tumor-Host Interactions in the B-Cell Lymphoma Microenvironment-Fueling Progression and Novel Brakes for Therapy. Int. J. Mol. Sci. 2019, 20, 4158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  440. Li, X.; Wenes, M.; Romero, P.; Huang, S.C.; Fendt, S.M.; Ho, P.C. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 425–441. [Google Scholar] [CrossRef]
  441. Xu, X.; Gnanaprakasam, J.N.R.; Sherman, J.; Wang, R. A Metabolism Toolbox for CAR T Therapy. Front. Oncol. 2019, 9, 322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  442. Xia, X.; Zhou, W.; Guo, C.; Fu, Z.; Zhu, L.; Li, P.; Xu, Y.; Zheng, L.; Zhang, H.; Shan, C.; et al. Glutaminolysis Mediated by MALT1 Protease Activity Facilitates PD-L1 Expression on ABC-DLBCL Cells and Contributes to Their Immune Evasion. Front. Oncol. 2018, 8, 632. [Google Scholar] [CrossRef]
  443. Blombery, P. Mechanisms of intrinsic and acquired resistance to venetoclax in B-cell lymphoproliferative disease. Leuk. Lymphoma 2020, 61, 257–262. [Google Scholar] [CrossRef]
  444. Diepstraten, S.T.; Anderson, M.A.; Czabotar, P.E.; Lessene, G.; Strasser, A.; Kelly, G.L. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat. Rev. Cancer 2022, 22, 45–64. [Google Scholar] [CrossRef]
  445. Lipsky, A.; Lamanna, N. Managing toxicities of Bruton tyrosine kinase inhibitors. Hematol. Am. Soc. Hematol. Educ. Program 2020, 2020, 336–345. [Google Scholar] [CrossRef] [PubMed]
  446. George, B.; Chowdhury, S.M.; Hart, A.; Sircar, A.; Singh, S.K.; Nath, U.K.; Mamgain, M.; Singhal, N.K.; Sehgal, L.; Jain, N. Ibrutinib Resistance Mechanisms and Treatment Strategies for B-Cell lymphomas. Cancers 2020, 12, 1328. [Google Scholar] [CrossRef] [PubMed]
  447. Wright, S.C.E.; Vasilevski, N.; Serra, V.; Rodon, J.; Eichhorn, P.J.A. Mechanisms of Resistance to PI3K Inhibitors in Cancer: Adaptive Responses, Drug Tolerance and Cellular Plasticity. Cancers 2021, 13, 1538. [Google Scholar] [CrossRef] [PubMed]
  448. Yan, W.; Liu, Z.; Liu, J.; Xia, Y.; Hu, K.; Yu, J. Application of Chimeric Antigen Receptor T Cells in the Treatment of Hematological Malignancies. Biomed. Res. Int. 2020, 2020, 4241864. [Google Scholar] [CrossRef] [PubMed]
  449. Edeline, J.; Houot, R.; Marabelle, A.; Alcantara, M. CAR-T cells and BiTEs in solid tumors: Challenges and perspectives. J. Hematol. Oncol. 2021, 14, 65. [Google Scholar] [CrossRef]
  450. Choi, B.D.; Yu, X.; Castano, A.P.; Bouffard, A.A.; Schmidts, A.; Larson, R.C.; Bailey, S.R.; Boroughs, A.C.; Frigault, M.J.; Leick, M.B.; et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat. Biotechnol. 2019, 37, 1049–1058. [Google Scholar] [CrossRef]
  451. Yin, Y.; Rodriguez, J.L.; Li, N.; Thokala, R.; Nasrallah, M.P.; Hu, L.; Zhang, L.; Zhang, J.V.; Logun, M.T.; Kainth, D.; et al. Locally secreted BiTEs complement CAR T cells by enhancing killing of antigen heterogeneous solid tumors. Mol. Ther. 2022, 30, 2537–2553. [Google Scholar] [CrossRef] [PubMed]
  452. Ben-Shmuel, A.; Biber, G.; Barda-Saad, M. Unleashing Natural Killer Cells in the Tumor Microenvironment-The Next Generation of Immunotherapy? Front. Immunol. 2020, 11, 275. [Google Scholar] [CrossRef]
  453. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. eBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef]
  454. Islam, R.; Pupovac, A.; Evtimov, V.; Boyd, N.; Shu, R.; Boyd, R.; Trounson, A. Enhancing a Natural Killer: Modification of NK Cells for Cancer Immunotherapy. Cells 2021, 10, 1058. [Google Scholar] [CrossRef] [PubMed]
  455. Berenson, J.R.; Martinez, D.; Safaie, T.; Silagan, N.; To, J.; Spektor, T.M.; Forouzan, E.; Swift, R.; Eades, B.; Eshaghian, S.; et al. Efficacy and Safety of Ruxolitinib and Steroids for Treating Patients with Relapsed or Refractory Multiple Myeloma. Blood 2021, 138, 2727. [Google Scholar] [CrossRef]
  456. Italiano, A.; Soria, J.C.; Toulmonde, M.; Michot, J.M.; Lucchesi, C.; Varga, A.; Coindre, J.M.; Blakemore, S.J.; Clawson, A.; Suttle, B.; et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: A first-in-human, open-label, phase 1 study. Lancet Oncol. 2018, 19, 649–659. [Google Scholar] [CrossRef] [PubMed]
  457. Sarkozy, C.; Morschhauser, F.; Dubois, S.; Molina, T.; Michot, J.M.; Cullières-Dartigues, P.; Suttle, B.; Karlin, L.; Le Gouill, S.; Picquenot, J.M.; et al. A LYSA Phase Ib Study of Tazemetostat (EPZ-6438) plus R-CHOP in Patients with Newly Diagnosed Diffuse Large B-Cell Lymphoma (DLBCL) with Poor Prognosis Features. Clin. Cancer Res. 2020, 26, 3145–3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  458. Morschhauser, F.; Tilly, H.; Chaidos, A.; McKay, P.; Phillips, T.; Assouline, S.; Batlevi, C.L.; Campbell, P.; Ribrag, V.; Damaj, G.L.; et al. Tazemetostat for patients with relapsed or refractory follicular lymphoma: An open-label, single-arm, multicentre, phase 2 trial. Lancet Oncol. 2020, 21, 1433–1442. [Google Scholar] [CrossRef]
  459. Major, A.; Kline, J.; Karrison, T.G.; Fishkin, P.A.S.; Kimball, A.S.; Petrich, A.M.; Nattam, S.; Rao, K.; Sleckman, B.G.; Cohen, K.; et al. Phase I/II clinical trial of temsirolimus and lenalidomide in patients with relapsed and refractory lymphomas. Haematologica 2022, 107, 1608–1618. [Google Scholar] [CrossRef]
  460. Tan, F.H.; Putoczki, T.L.; Stylli, S.S.; Luwor, R.B. Ponatinib: A novel multi-tyrosine kinase inhibitor against human malignancies. Onco Targets Ther. 2019, 12, 635–645. [Google Scholar] [CrossRef] [Green Version]
  461. Cortes, J.E.; Kim, D.W.; Pinilla-Ibarz, J.; le Coutre, P.D.; Paquette, R.; Chuah, C.; Nicolini, F.E.; Apperley, J.F.; Khoury, H.J.; Talpaz, M.; et al. Ponatinib efficacy and safety in Philadelphia chromosome-positive leukemia: Final 5-year results of the phase 2 PACE trial. Blood 2018, 132, 393–404. [Google Scholar] [CrossRef]
  462. Martinelli, G.; Papayannidis, C.; Piciocchi, A.; Robustelli, V.; Soverini, S.; Terragna, C.; Marconi, G.; Lemoli, R.M.; Guolo, F.; Fornaro, A.; et al. INCB84344-201: Ponatinib and steroids in frontline therapy for unfit patients with Ph+ acute lymphoblastic leukemia. Blood Adv. 2022, 6, 1742–1753. [Google Scholar] [CrossRef]
  463. Ribera, J.M.; García-Calduch, O.; Ribera, J.; Montesinos, P.; Cano-Ferri, I.; Martínez, P.; Esteve, J.; Esteban, D.; García-Fortes, M.; Alonso, N.; et al. Ponatinib, chemotherapy, and transplant in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia. Blood Adv. 2022, 6, 5395–5402. [Google Scholar] [CrossRef]
  464. Short, N.J.; Kantarjian, H.M.; Konopleva, M.; Jain, N.; Huang, X.; Ravandi, F.; Wierda, W.G.; Borthakur, G.; Sasaki, K.; Issa, G.C.; et al. Combination of ponatinib and blinatumomab in Philadelphia chromosome-positive acute lymphoblastic leukemia: Early results from a phase II study. J. Clin. Oncol. 2021, 39, 7001. [Google Scholar] [CrossRef]
  465. Peterson, T.J.; Orozco, J.; Buege, M. Selinexor: A First-in-Class Nuclear Export Inhibitor for Management of Multiply Relapsed Multiple Myeloma. Ann. Pharmacother. 2020, 54, 577–582. [Google Scholar] [CrossRef] [PubMed]
  466. Sarnik, J.; Popławski, T.; Tokarz, P. BET Proteins as Attractive Targets for Cancer Therapeutics. Int. J. Mol. Sci. 2021, 22, 11102. [Google Scholar] [CrossRef]
  467. Burgoyne, A.M.; Vann, K.R.; Joshi, S.; Morales, G.A.; Vega, F.M.; Singh, A.; Pal, D.; Merati, A.B.; Kutateladze, T.G.; Durden, D.L. A triple action CDK4/6-PI3K-BET inhibitor with augmented cancer cell cytotoxicity. Cell Discov. 2020, 6, 49. [Google Scholar] [CrossRef] [PubMed]
  468. Smith, A.L.; Eiken, A.P.; Skupa, S.A.; Moore, D.Y.; Umeta, L.T.; Smith, L.M.; Lyden, E.R.; D’Angelo, C.R.; Kallam, A.; Vose, J.M.; et al. A Novel Triple-Action Inhibitor Targeting B-Cell Receptor Signaling and BRD4 Demonstrates Preclinical Activity in Chronic Lymphocytic Leukemia. Int. J. Mol. Sci. 2022, 23, 6712. [Google Scholar] [CrossRef] [PubMed]
  469. Vann, K.R.; Pal, D.; Smith, A.L.; Sahar, N.E.; Krishnaiah, M.; El-Gamal, D.; Kutateladze, T.G. Combinatorial inhibition of BTK, PI3K-AKT and BRD4-MYC as a strategy for treatment of mantle cell lymphoma. Mol. Biomed. 2022, 3, 2. [Google Scholar] [CrossRef]
  470. Martinez-Outschoorn, U.E.; Peiris-Pagés, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective. Nat. Rev. Clin. Oncol. 2017, 14, 113. [Google Scholar] [CrossRef]
Figure 1. Models for the signaling pathways regulated by BCL2 family proteins, BCR components, and TAAs (CD19, CD20, CD22, CD38) in B cell malignancies. (a) Prosurvival BCL2-like proteins (Bcl-2, Bcl-xL, Bcl-w, and Mcl-1) interact with the proapoptotic members called BH3-only proteins (Bim, Bid, Puma, Bad, and Noxa), or bind to the apoptotic effectors Bax and Bak and sequester them in an inactive form. The pro-apoptotic proteins induce apoptosis by activating Bax and Bak either directly or indirectly through inhibition of the anti-apoptotic proteins. Once activated, Bax and Bak form oligomers; this leads to pore formation, mitochondrial outer membrane permeabilization (MOMP), and then the release of cytochrome c and other pro-death proteins from mitochondria, resulting in caspase activation and cell apoptosis. (b) Upon BCR ligation and activation, Lyn phosphorylates CD79A/B, which activates SYK. In turn, activated SYK phosphorylates and recruits the B-cell linker protein (BLNK), which binds to BTK and PLCγ2 and catalyzes the cleavage of membrane phosphatidyl inositol bisphosphate into inositol triphosphate and diacyl glycerol. This releases Ca2+ from intracellular stores and activates PKCβ and downstream proteins. The positive BCR coreceptors CD19 and CD20 are phosphorylated by LYN during BCR signaling, leading to the recruitment of PI3K to the BCR. Together, these signaling pathways activate the ERK, NF-κB, AKT/mTOR pathways. The negative B cell coreceptor CD22 is phosphorylated by LYN and inhibits the BCR signal by recruiting SHP-1, which dephosphorylates and inactivates BLNK and CD19 and thus leads to a decrease in the cytoplasmic Ca2+ concentration. SHP-1 also decreases the cytoplasmic Ca2+ concentration by activating the plasma membrane Ca2+-ATPase (PMCA4) and thus promoting Ca2+ efflux. CD38 activation leads to the phosphorylation of ZAP70 and further sustains the signal mediated by the BCR. (c) Independently of the BCR signaling pathway, CD19, CD20, CD22, and CD38 have a role in activating proliferation/survival pathways in malignant B cells. CD38 (via ZAP70 activation), CD19 and CD20 are linked to the proliferation/survival signaling pathways controlled by CXCR4. Through CD38′s enzymatic activity, the reaction products (nicotinamide adenine dinucleotide, cyclic ADP-ribose, and ADP-ribose) are used inside the cells to open different Ca2+ stores, which leads to an increase in the cytoplasmic Ca2+ concentration independently of the conventional IP3 pathway. CD22 ligation activates AP-1, c-jun, and the c-jun NH2-terminal kinases (JNKs).
Figure 1. Models for the signaling pathways regulated by BCL2 family proteins, BCR components, and TAAs (CD19, CD20, CD22, CD38) in B cell malignancies. (a) Prosurvival BCL2-like proteins (Bcl-2, Bcl-xL, Bcl-w, and Mcl-1) interact with the proapoptotic members called BH3-only proteins (Bim, Bid, Puma, Bad, and Noxa), or bind to the apoptotic effectors Bax and Bak and sequester them in an inactive form. The pro-apoptotic proteins induce apoptosis by activating Bax and Bak either directly or indirectly through inhibition of the anti-apoptotic proteins. Once activated, Bax and Bak form oligomers; this leads to pore formation, mitochondrial outer membrane permeabilization (MOMP), and then the release of cytochrome c and other pro-death proteins from mitochondria, resulting in caspase activation and cell apoptosis. (b) Upon BCR ligation and activation, Lyn phosphorylates CD79A/B, which activates SYK. In turn, activated SYK phosphorylates and recruits the B-cell linker protein (BLNK), which binds to BTK and PLCγ2 and catalyzes the cleavage of membrane phosphatidyl inositol bisphosphate into inositol triphosphate and diacyl glycerol. This releases Ca2+ from intracellular stores and activates PKCβ and downstream proteins. The positive BCR coreceptors CD19 and CD20 are phosphorylated by LYN during BCR signaling, leading to the recruitment of PI3K to the BCR. Together, these signaling pathways activate the ERK, NF-κB, AKT/mTOR pathways. The negative B cell coreceptor CD22 is phosphorylated by LYN and inhibits the BCR signal by recruiting SHP-1, which dephosphorylates and inactivates BLNK and CD19 and thus leads to a decrease in the cytoplasmic Ca2+ concentration. SHP-1 also decreases the cytoplasmic Ca2+ concentration by activating the plasma membrane Ca2+-ATPase (PMCA4) and thus promoting Ca2+ efflux. CD38 activation leads to the phosphorylation of ZAP70 and further sustains the signal mediated by the BCR. (c) Independently of the BCR signaling pathway, CD19, CD20, CD22, and CD38 have a role in activating proliferation/survival pathways in malignant B cells. CD38 (via ZAP70 activation), CD19 and CD20 are linked to the proliferation/survival signaling pathways controlled by CXCR4. Through CD38′s enzymatic activity, the reaction products (nicotinamide adenine dinucleotide, cyclic ADP-ribose, and ADP-ribose) are used inside the cells to open different Ca2+ stores, which leads to an increase in the cytoplasmic Ca2+ concentration independently of the conventional IP3 pathway. CD22 ligation activates AP-1, c-jun, and the c-jun NH2-terminal kinases (JNKs).
Cancers 14 06026 g001
Figure 2. Timeline from 1997 onwards, showing the development of therapeutics for B cell malignancies. (a) (1997–2020) Drugs currently approved by the FDA for the treatment of B cell malignancies; (b) investigational drugs in clinical development (2013–2021) for B cell malignancies. ADC, antibody–drug conjugate; BCL2, B-cell lymphoma/leukemia 2; BiTE, bispecific T-cell engager, BTK, Bruton tyrosine kinase; CAR-T, chimeric antigen receptor-T cells; PI3K, phosphatidylinositol-3 kinase.
Figure 2. Timeline from 1997 onwards, showing the development of therapeutics for B cell malignancies. (a) (1997–2020) Drugs currently approved by the FDA for the treatment of B cell malignancies; (b) investigational drugs in clinical development (2013–2021) for B cell malignancies. ADC, antibody–drug conjugate; BCL2, B-cell lymphoma/leukemia 2; BiTE, bispecific T-cell engager, BTK, Bruton tyrosine kinase; CAR-T, chimeric antigen receptor-T cells; PI3K, phosphatidylinositol-3 kinase.
Cancers 14 06026 g002aCancers 14 06026 g002b
Figure 3. The cell’s main metabolic pathways and the corresponding inhibitors. Glucose, free fatty acids (FFAs) and glutamine enter cells via glucose transporters (GLUT), CD36, and solute carrier (SLC) transporters respectively. Once inside the cells, these substrates are catabolized by various metabolic reactions in the cytosol and the mitochondria in the tricarboxylic acid cycle (TCA). Cofactors produced by the TCA are used by the electron transport chain (ETC) to produce ATP. Some cancer cells shift their metabolism (and specifically glycolysis) towards lactate production; ATP is generated outside the mitochondria. Lactate can then be exported from the cells via monocarboxylate transporters (MCTs). Inhibitors are shown in red, key enzymes are labeled in green, and metabolic pathways are shown in pink. αKG, α-ketoglutaric acid; FASN, fatty acid synthase; FAO, fatty acid oxidation; GLS, glutaminase; HK2, hexokinase 2; LDHA, lactate dehydrogenase A; PDH, pyruvate dehydrogenase; PDK4, pyruvate dehydrogenase kinase 4.
Figure 3. The cell’s main metabolic pathways and the corresponding inhibitors. Glucose, free fatty acids (FFAs) and glutamine enter cells via glucose transporters (GLUT), CD36, and solute carrier (SLC) transporters respectively. Once inside the cells, these substrates are catabolized by various metabolic reactions in the cytosol and the mitochondria in the tricarboxylic acid cycle (TCA). Cofactors produced by the TCA are used by the electron transport chain (ETC) to produce ATP. Some cancer cells shift their metabolism (and specifically glycolysis) towards lactate production; ATP is generated outside the mitochondria. Lactate can then be exported from the cells via monocarboxylate transporters (MCTs). Inhibitors are shown in red, key enzymes are labeled in green, and metabolic pathways are shown in pink. αKG, α-ketoglutaric acid; FASN, fatty acid synthase; FAO, fatty acid oxidation; GLS, glutaminase; HK2, hexokinase 2; LDHA, lactate dehydrogenase A; PDH, pyruvate dehydrogenase; PDK4, pyruvate dehydrogenase kinase 4.
Cancers 14 06026 g003
Table 4. Selected ongoing and now-recruiting clinical trials of novel anti-TAA inhibitors in B cell malignancies.
Table 4. Selected ongoing and now-recruiting clinical trials of novel anti-TAA inhibitors in B cell malignancies.
DrugClassDisease SettingPhaseStudy References
Belantamab
mafodotin/
GSK2857916
ADC anti-BCMA-monomethyl
auristatin F
R/R MMI/II






I/II

NCT03848845
Active, 2018-
Alone or combined with pembrolizumab

NCT04126200
NCT03544281
NCT03715478
Recruiting, 2018-
Alone or combined with current therapies
[273]








Teclistamab/
JNJ-64007957
BiTE anti-BCMA/CD3 R/R MMI/II




I





III
NCT04557098
Active, 2017–2024


NCT04586426
Recruiting, 2019–2024
Combined with talquetamab or daratumuab or pomalidomide

NCT05083169
Recruiting, 2021–2026
daratumumab compared with current therapies
[278]
Elranatamab/
PF-06863135
BiTE anti-BCMA/CD3 Refractory MM IINCT04649359
Active, 2020–2024
Idecabtagene vicleucel/
bb2121
Anti-BCMA CAR-T R/R MMIINCT03361748
Active, 2017–2024
[279]
BM38Dual anti-BCMA/CD38 CAR-TR/R MMIChiCTR1900026286
Active, 2019-
[281]
BCMA-CAR-NK92Anti-BCMA CAR-NKR/R MMI/IINCT03940833
Recruiting, 2019–2022
[282]
Lanalumab/VAY-736Anti-BAFF-R Ab CLLINCT03400176,
Recruiting, 2018-
Alone or combined with ibrutinib
BAFFR-CAR TAnti-BAFF-R CAR-TB-ALL, R/R MCLINCT04690595, NCT05370430
Recruiting, 2020–2024
Cirmtuzumab/
UC961
newly renamed
Zilovertamab
Anti-ROR1 AbCLL




CLL, SLL, MCL




R/R MCL
II




I/II




III
NCT04501939
Recruiting, 2020-
Combined with venetoclax

NCT03088878
Active, 2018–2025
Combined with ibrutinib

NCT05431179
Recruiting, 2022-
Combined with ibrutinib
Pembrolizumab/MK-3475Anti-PD1 Ab recruiting immune cells to attack tumor B cellsR/R CLL, B-NHLIINCT02332980
Active, 2015–2023
Alone or combined with ibrutinib or idelalisib
[283]
High-Risk or R/R CLL/SLL II NCT03204188
Active, 2017–2030
Combined with ibrutinib, fludarabine
R/R WM II NCT03630042
Recruiting, 2018-
Combined with rituximab
R/R B-ALL I/II NCT03512405
Recruiting, 2018-
Combined with blinatumomab
[284]
R/R DLBCL I NCT03340766
Active, 2017-
Combined with blinatumomab
Cemiplimab/
REGN2810









Anti-PD1 Ab recruiting immune cells to attack tumor B cells








B-NHL




R/R MM



DLBCL


I




I/II



I/II


NCT02651662
Active, 2016-
Combined with REGN1979 (anti-CD20-CD3)

NCT03194867
Active, 2016-
Combined with isatuximab

NCT03769181
Active, 2018-
Combined with isatuximab










[285]

AtezolizumabAnti-PD-L1 AbDLBCLI/IINCT02926833
Active, 2016–2022
Alone or combined with axicabtagene ciloleucel
[286]
Ulocuplumab/
BMS-936564
Anti-CXCR4 AbWMI/IINCT03225716
Active, 2017–2025
Alone or combined with ibrutinib
[287]
Mavorixafor/
AMD070
CXCR4 antagonist (blocks CXCL12 binding)WMINCT04274738
Active, 2020–2025
Alone or combined with
ibrutinib
CS1-CAR-TCS1-CAR-T/IL7-
CCL19
R/R MMINCT03778346
Recruiting, 2018–2022
CS1-CAR-TCS1-CAR-T/41BB-
tEGFR
R/R MMINCT03710421
Recruiting, 2018–2023
following chemotherapy
J1/MelflufenMelphalan-p-fluoro-L-phenylalalnine (aminopeptidase substrat)MM refractory to lenalidomideIIINCT03151811
Active, 2017-
combined with dexamethasone compared to pomalidomide + dexamethasone
[288]
Brentuximab vedotinADC anti-CD30-mono methyl auristatin ER/R B-NHLIINCT03646123
Active, 2018–2026
Lilotomab/
tetulomab/
betalutin
ARC anti-CD37-177lutetium Relapsed B-NHLI/IINCT01796171
Recruiting, 2013–2026
[289]
GEN3009DuoHexaBody-CD37R/R B-NHLINCT04358458
Recruiting, 2020–2025
CART30Anti-CD30 CAR-TB-NHLINCT02259556
Recruiting, 2016–2029
Combined with cyclophosphamide and fludarabine
[290]
CD30.CAR-TsAnti-CD30 CAR-TB-NHLI/IINCT02690545
Recruiting, 2016–2038
Combined with fludarabine and bendamustine
[291]
CD30.CAR TAnti-CD30 CAR-TB-NHLINCT02917083
Recruiting, 2018–2036
ICAR30Anti-CD30 CAR-TB-NHLINCT03383965
Recruiting, 2017–2025
ATLCAR.CD30 Anti-CD30 CAR-TB-NHLINCT026663297
Active, 2016–2037
Magrolimab/
Hu5F9-G4






Anti-CD47






R/R B-NHLI/IINCT02953509
Active, 2016–2026
Combined with rituximab and (in some cases) with chemotherapy
[292]
R/R MM II NCT04892446
Recruiting, 2021–2024
Combined with daratumumab, or pomalidomide and dexamethasone, or bortezomib and dexamethasone
R/R B-NHL II NCT04788043
Recruiting, 2021–2027
Combined with pembrolizumab
IBI322Bi anti-CD47/PD-L1ALL, CLL/SLL B-NHLINCT04795128
Recruiting, 2021–2024
TG-1801/
NI-1701
Bi anti-CD47/CD19B-NHL




B-NHL, CLL
I




Ib
NCT03804996
Active, 2019–2022
Combined with ublituximab

NCT04806035
Recruiting, 2021–2023
Alone or combined with ublituximab



[293]
CPO107/
JMT601
Fusion protein Anti-CD20-SIRPαAdvanced
B-NHL
INCT04853329
Recruiting, 2021–2024
STRO-001ADC anti-CD74-
maytansoid
DLBCL, FL, MMINCT03424603
Recruiting, 2018–2023
[294]
Polatuzumab vedotinADC anti-CD79b-monomethyl auristatin EDLBCLINCT04231877
Recruiting, 2020–2031
Combined with rituximab, prednisone, etoposide, doxorubicin, cyclophosphamide
R/R DLBCLI NCT04739813
Recruiting, 2021–2027
Combined with venetoclax, ibrutinib, prednisone, obinutuzumab, and lenalidomide
R/R B-NHLIb/II NCT03533283
Recruiting, 2018–2024
Combined with glofitamab
Cevostomab/
BFCR4350A
BiTE-anti-FcRL5/CD3 R/R MMINCT03275103
Recruiting, 2017–2023
[295]
Talquetamab/
JNJ-64407564
BiTE duoBody-anti-GPRC5D/CD3 R/R MMI/IINCT03399799
Recruiting, 2018–2025
[296]
ADAM17, a disintegrin and metalloprotease 17; ADC, antibody drug conjugate; BAFF-R, B-cell activating factor receptor; BCMA, B cell maturation antigen FcRL5, Fc receptor-like 5; GPRC5D, G-protein coupled receptor family C group 5 member D; ROR1, receptor tyrosine kinase-like orphan receptor 1; tEGFR, truncated epidermal growth factor receptor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tannoury, M.; Garnier, D.; Susin, S.A.; Bauvois, B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What’s Coming Next? Cancers 2022, 14, 6026. https://doi.org/10.3390/cancers14246026

AMA Style

Tannoury M, Garnier D, Susin SA, Bauvois B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What’s Coming Next? Cancers. 2022; 14(24):6026. https://doi.org/10.3390/cancers14246026

Chicago/Turabian Style

Tannoury, Mariana, Delphine Garnier, Santos A. Susin, and Brigitte Bauvois. 2022. "Current Status of Novel Agents for the Treatment of B Cell Malignancies: What’s Coming Next?" Cancers 14, no. 24: 6026. https://doi.org/10.3390/cancers14246026

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop