Next Article in Journal
Molecular Biology of Pediatric and Adult Ovarian Germ Cell Tumors: A Review
Next Article in Special Issue
Heterogeneity in the Metastatic Microenvironment: JunB-Expressing Microglia Cells as Potential Drivers of Melanoma Brain Metastasis Progression
Previous Article in Journal
Deciphering the Role of the Anaphylatoxin C3a: A Key Function in Modulating the Tumor Microenvironment
Previous Article in Special Issue
Acidic Growth Conditions Promote Epithelial-to-Mesenchymal Transition to Select More Aggressive PDAC Cell Phenotypes In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice

by
Anna Chen
,
Ines Neuwirth
and
Dietmar Herndler-Brandstetter
*
Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, 1090 Vienna, Austria
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(11), 2989; https://doi.org/10.3390/cancers15112989
Submission received: 13 March 2023 / Revised: 10 May 2023 / Accepted: 28 May 2023 / Published: 30 May 2023
(This article belongs to the Special Issue Microenvironment and Cancer Progression 2.0)

Abstract

:

Simple Summary

A bottleneck in oncology is the translation of results from preclinical models to the clinics. The rate of anticancer drugs that are effective in preclinical studies but fail in clinical trials is more than 95%. In order to test new immunotherapies and to identify the most effective combination of anticancer drugs, next-generation mouse models have been developed. These “humanized mouse models” support the growth of patient-derived tumors and the development of a human immune system. This review provides an overview of next-generation humanized mouse models and how they can be used to advance precision cancer medicine and immuno-oncology clinical trial design.

Abstract

Cancer immunotherapy has brought significant clinical benefits to numerous patients with malignant disease. However, only a fraction of patients experiences complete and durable responses to currently available immunotherapies. This highlights the need for more effective immunotherapies, combination treatments and predictive biomarkers. The molecular properties of a tumor, intratumor heterogeneity and the tumor immune microenvironment decisively shape tumor evolution, metastasis and therapy resistance and are therefore key targets for precision cancer medicine. Humanized mice that support the engraftment of patient-derived tumors and recapitulate the human tumor immune microenvironment of patients represent a promising preclinical model to address fundamental questions in precision immuno-oncology and cancer immunotherapy. In this review, we provide an overview of next-generation humanized mouse models suitable for the establishment and study of patient-derived tumors. Furthermore, we discuss the opportunities and challenges of modeling the tumor immune microenvironment and testing a variety of immunotherapeutic approaches using human immune system mouse models.

1. Introduction

Humanized mice are powerful models for studying human biology, disease and therapeutic interventions. By definition, humanized mice are animals that have either been xenografted with human cells or genetically modified to express human genes. Humanized mouse models aim to more faithfully recapitulate important features of human biology and disease, and therefore represent an important pillar of translational biomedical research and precision medicine-based approaches. In the past 20 years, humanized mice have been used to study the human immune system and infectious diseases (e.g., human immunodeficiency virus (HIV) [1,2], coronavirus disease 2019 (COVID-19) [3,4], hepatitis B and C viruses [5,6], Epstein-Barr virus (EBV) [7] and dengue virus [8]), human erythropoiesis and sickle cell disease [9], malignant diseases (e.g., leukemia [10,11,12], lymphoma [13,14], multiple myeloma [15,16], myelodysplastic syndrome [17], melanoma [18,19]), as well as therapeutic interventions, including cancer immunotherapy [20] and chimeric antigen receptor (CAR) T cell therapy [21].
Although immunotherapy is becoming a cornerstone of modern oncology, complete and durable responses are only observed in a small fraction of cancer patients. For example, results from clinical trials using anti-PD-1 and anti-CTLA-4 checkpoint inhibitors indicate that clinical benefit is limited to only 5% of metastatic colorectal cancer (mCRC) patients who have heavily mutated tumors and tumor-infiltrating T cells [22]. In contrast, 95% of mCRC patients who have a mismatch-repair-proficient, microsatellite instability-low (pMMR/MSI-L) phenotype did not show any clinical benefit. To make matters worse, the success rate of anticancer drugs, i.e., drugs that are effective in preclinical studies as well as clinical trials, is estimated at only 3.4% compared to 20.9% for drugs in all other therapeutic areas of medicine [23]. Hence, there is not only a need for new immunotherapies and more predictive biomarkers, but also preclinical models that accurately reflect the patient’s tumor heterogeneity and tumor immune microenvironment (TIME) [24]. In view of the recent development of a plethora of new immunotherapeutic drugs, it will become more difficult to recruit sufficient numbers of patients to validate all potentially effective combination immunotherapies in clinical trials. Next-generation patient-derived tumor xenograft (PDX) humanized mouse models could be used for preclinical evaluation of the efficacy of combination (immuno)therapies and treatment regimens, for identifying biomarkers of responsiveness, and to stratify cancer patients for clinical trials.
Immunocompromised mice are widely used for human tumor xenotransplantation. In 1966, nude mice that lack the majority of mouse T cells were generated [25]. In 1983, BALB/c-Ighb (C.B-17) mice with a spontaneous mutation in the Prkdc gene (Prkdcscid) were discovered [26]. These mice lacked mouse T and B cells and were called severe combined immunodeficiency (SCID) mice. Backcrossing SCID mice onto a non-obese diabetic (NOD) background (NOD-SCID) resulted in an impaired function of mouse natural killer (NK) cells and a mutation in signal regulatory protein alpha (SIRPA), which conferred high affinity to human CD47 and therefore protected human immune cells from phagocytosis by mouse myeloid cells [27]. However, NOD-SCID mice showed residual NK cell activity and developed lymphomas in old age. To improve PDX engraftment and human immune cell development, NOD-SCID mice with a knockout of the common gamma chain (IL2rg) were generated in 2002 (NSG) [28,29]. Since then, similar highly immunodeficient first-generation humanized mouse strains have been developed, such as NOG [30], BRG [7] and NRG [31] (Figure 1). Similar to the Prkdcscid mutation, knock-out of Rag1 (NRG) or Rag2 (BRG) led to a loss of T and B cells [32]. However, humanized mice on a NOD background (NSG, NOG, NRG) allowed better hematopoietic engraftment compared to other strains, such as C57BL/6 (B6RG) or BALB/c (BRG). This was due to a polymorphism in SIRPA, since SIRPANOD demonstrated enhanced binding to the human CD47 ligand [33]. The expression of human SIRPA (SRG) or SIRPANOD (BRGS) (Figure 1) was required to prevent phagocytosis of human cells by mouse macrophages and thereby improved human immune cell reconstitution and long-term survival of human immune cell lineages. Providing this “do not eat me signal” in SRG and BRGS mice resulted in human immune system engraftment comparable to that in NSG and NRG mice [14,34,35].
The highly immunodeficient mouse strains NSG, NOG, NRG, BRGS and SRG support the growth of PDXs as well as the development of a human immune system following engraftment with human CD34+ hematopoietic stem and progenitor cells (HSPCs). To further improve HSPC and/or PDX engraftment in these mice, numerous genetic modifications have been added to first-generation humanized mouse strains. A genealogical tree of these novel humanized mouse strains is shown in Figure 1. These next-generation humanized mouse models may (1) be more immunodeficient, as they not only lack mouse T, B and NK cells but also have defects in mouse myeloid cell development (e.g., NRG-F, BRGS-F, MISTRG and NOG-GCSF) and/or (2) express human genes in order to promote the growth of PDXs or to improve the development of a diverse and functional human immune system (e.g., MISTRG, NSG-SGM3, NRG-SGM3, NOG-EXL and BRGS-T) [36]. Next-generation humanized mice have been generated to further improve the development and function of human immune cell lineages, thereby enabling preclinical testing of different immunotherapeutic agents. Technological advances, such as CRISPR/Cas9-mediated genome engineering, have facilitated the development of novel next-generation humanized mouse strains, including targeted genomic humanization of mice, so called “knock-in” humanized mice [37,38]. To support human T cell development, humanized mice have been developed that express (1) human cytokines, such as interleukin (IL)-7 (NSG-W41-IL7 [39], NSG-IL7-IL15 [40]), IL-15 (SRG-15 [14], NSG-IL15 [41], NSG-IL7-IL15 [40], NOG-IL15 [42]) or thymic stromal lymphopoietin (TSLP; BRGST mice) [43]) or (2) HLA class I and II molecules, such as HLA-A2 (NSG-A2 [44], NOG-A2 [45], NRG-A2/DR4 [46], BRGS-F-A2 [47], BRGS-A2/DR2 [48]), HLA-DR1 (NSG-A2/DR1) [49], HLA-DR2 (BRGS-A2/DR2) [48], HLA-DR4 (NRG-A2/DR4 [46], NOG-DR4 [50]) and HLA-DQ8 (NSG-DQ8) [51] (Figure 1).
This review summarizes next-generation humanized mouse models used to establish PDXs of various cancer types and discusses their utility for preclinical testing of different immunotherapeutic strategies and combination treatments to guide clinical trial design.

2. Establishing and Modeling Patient-Derived Tumors in Humanized Mice

Cell line-derived xenograft (CDX) models using tumor cells immortalized in vitro from patient tissues have been widely used for preclinical drug development. Tumor cell lines provide an infinite source of biological material, the underlying genetic abnormalities are usually well characterized, and they can be easily cultured and genetically manipulated. However, CDX models often do not accurately mimic tumor heterogeneity and are poorly predictive of efficacy in phase III clinical trials, as evidenced by the high rates of drug attrition in cancer [23,52,53]. Because of the low success rate in establishing cancer cell lines, i.e., only ~10% for colorectal cancer (CRC) [54] or pancreatic cancer [55], the field of oncology has relied on a very limited number of human CDXs per cancer type for preclinical testing of anticancer drugs.
PDXs are generated by transplantation of patient tumors (single cell suspension or tissue fragment) into immunodeficient mice and in vivo propagation for at least 3–5 passages (Figure 2A). PDX models are considered to better recapitulate the histological features, molecular characteristics and intratumor heterogeneity of human cancers, thereby overcoming some of the limitations of CDX models [17,56,57,58,59]. However, establishing PDXs can take anywhere from several months to more than a year, and depends on a variety of factors, such as the cancer (sub)type, the quantity and quality of the tumor sample, the implantation technique, the implantation site, the immunodeficient mouse strain as well as supplementation of human factors (e.g., hormones for hormone receptor-positive cancers) (Figure 2B) [60,61,62]. In general, metastases engraft better than primary tumors [63] and the more immunodeficient the mouse strain, the greater the likelihood that the patient tumor will engraft and reach stable tumor growth after 5 in vivo passages [64,65,66]. Yet, successful establishment of PDXs has also been achieved when using nude mice that have a lower level of immunodeficiency compared to NSG mice [61,67,68]. Because the mouse host may affect tumor evolution during PDX engraftment and propagation, several studies have extensively analyzed copy number alterations (CNAs) in different PDX models. Guillen and colleagues showed that common driver mutations in breast cancer PDXs were retained in early and late passages with minimal CNAs [69]. By comparing a larger collection of 1127 PDX samples and their originating 324 patient tumors, Woo et al. demonstrated that CNA profiles were conserved during engraftment and passaging of PDXs using CNA inferences based on DNA sequencing [70]. Some CNAs can, however, occur over long-term passaging (e.g., ≥18 passages), and large CNA discordances have even been observed in early passage PDXs, although only in 2.44% of samples [70]. The rare occurrence of large CNA changes suggests that variations observed in PDXs are mostly due to spontaneous selection of tumor clones, rather than a systematic selection pressure by the mouse environment. Another aspect of serial transplantation is the exchange of human stroma by mouse stroma in late passage PDXs, which can affect tumor growth and drug distribution. For instance, increased growth of late versus early passage PDXs has been reported, which may be due in part to the loss of human stroma cells in late passage PDXs [60]. Overcoming these limitations by minimizing passage numbers (a maximum of 5–7 passages has been recommended) [63,71], monitoring CNAs and clonal tumor heterogeneity as well as confirming expected molecular targets is critical to the reproducibility and translatability of preclinical drug screens in PDX mouse models.
Academic and industry institutions (e.g., EurOPDX, PDXNet, PRoXe, NCI-PDMR, PIVOT, COG xenograft repository, LIMORE, MURAL, NIBR PDXE) have established and characterized thousands of PDX models generated from a variety of cancer types, including CRC [61,63,72,73], breast cancer [56,69,74,75], esophagus carcinoma [73], hepatocellular carcinoma [57,62], melanoma [63,76], lung cancer [63,77], prostate cancer [78], gastric cancers [63], and leukemia and lymphoma [79] (Table 1). The success rate of establishing PDXs varies greatly between different tumor types. For example, high success rates of establishing PDXs have been reported for CRC (52–91%), pancreatic cancer (54–100%) and skin melanoma (62–90%) [63,76] whereas lower engraftment rates have been reported for breast cancer (4–86%) and prostate cancer (20.6%) [63,69,78]. In particular, hormone-dependent cancers such as breast and prostate cancers have been shown to have a very low engraftment rate. Accordingly, the huge variability in breast cancer engraftment rate was due to the different types of breast cancer. Engraftment rate for primary estrogen receptor positive (ER+) breast cancers was only 4–20% [63] whereas the engraftment rate for primary triple-negative breast cancer (TNBC) was 30–34% when transplanted subcutaneously, and 60–86% when transplanted orthotopically [63]. In general, orthotopic transplantation of patient tumors yielded higher engraftment rates compared to subcutaneous transplantation [63,80], and primary tumors showed a lower engraftment rate (CRC: 52–75%; TNBC: 30–34%) compared to metastases (CRC: 73–91%; TNBC: 60%) [63]. Of note, the human IL-2 in NOG-IL2 mice may activate tumor-resident T cells in transplanted tumor pieces and reduce the engraftment rate of PDXs in NOG-IL2 compared to NOG mice [81].
Several types of cancer rely on human factors for their growth. For instance, ER+ breast cancers have a very low rate of engraftment and expansion, which is enhanced when estradiol supplementation is provided [74,82,83]. By analyzing 62 TNBCs, Petrosyan et al. found that immunologically “cold” TNBCs engraft at a higher rate than immune cell-enriched “hot” TNBCs [84]. Thus, tumor-infiltrating human immune cells may affect the engraftment rate of some patient-derived tumors, although the mechanisms still need to be experimentally validated. Some human hematological neoplasms, i.e., certain types of leukemia and multiple myeloma, also show low engraftment in immunodeficient mice. For example, the reproducible generation of human acute myeloid leukemia (AML) xenografts is primarily limited to very aggressive “high-risk” cases. Less aggressive, so-called “favorable-risk” AML, which constitute approximately 40% of all AML cases, engraft poorly in immunodeficient mice [85]. Among those favorable-risk AML cases, inv(16) and isolated nucleophosmin mutation (NPM1mut) AMLs engrafted with high efficacy in MISTRG but not NSG mice [11] (Figure 2C). Human cytokines, in particular human macrophage colony-stimulating factor (M-CSF), are required for the efficient engraftment of inv(16) AML. NSG-SGM3 mice, also called NSGS mice, express human stem cell factor (SCF), granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin 3 (IL-3), and improve the engraftment rate of AMLs from 50% in NSG to 82% in NSG-SGM3 mice [86]. This allowed the identification of different patterns of relapse of AML caused by leukemia stem cells [87]. Improved engraftment of leukemic human hematopoietic cells, acute promyelocytic leukemia (APL) and myelofibrosis samples was achieved by using a humanized bone marrow ossicle xenotransplantation model [88]. Another hematologic malignancy, myelodysplastic syndromes (MDS), has been difficult to engraft in NSG mice, but CD34+ HSPCs from MDS patients engrafted well in MISTRG mice [17] (Table 1). Although transient engraftment was observed in NSG-SGM3 mice when HSPCs from MDS patients were co-injected with mesenchymal stromal cells [89], sustained engraftment of MDS in NSG-SGM3 was not observed when using MDS samples from 45 patients [86,90]. This is not surprising, since hematopoietic stem cell maintenance has been shown to be impaired in NSG-SGM3 mice [91,92]. It remains to be seen whether NSG-Quad mice, which are NSG-SGM3 mice that additionally express human M-CSF, will be as suitable a model as MISTRG (Figure 1) [93]. Multiple myeloma (MM) is a hematological neoplasia originating from malignant plasma cells in the bone marrow. Because these malignant plasma cells depend on human IL-6, high engraftment of MM has been achieved in MISTRG-6 humanized mice [16]. Clonal heterogeneity and evolution have major implications for disease progression and relapse in hematologic malignancies. Thus, it will be of great importance to choose humanized mouse models that produce the human factors relevant for the respective PDX entity to avoid animal model-driven changes in tumor clonality and thus biases in preclinical drug development [94,95].
Table 1. A selection of immunodeficient mouse models used to generate PDXs.
Table 1. A selection of immunodeficient mouse models used to generate PDXs.
Mouse ModelType of PDXNumber of
Patients
PDX
Engraftment Rate
References
NudeCRC8563.5%[61]
NOD-SCID, nudeCRC4871–74%[96]
SCIDUveal melanoma9028%[97]
NOD-SCIDCervical cancer2230.7%[98]
CRC (metastatic)8587%[99]
Esophageal squamous cell carcinoma2513.3%[100]
Gastric cancer18534.1%[101]
Leukemia (T-ALL)1952%[102]
Liposarcoma5644.6%[103]
NSCLC7549%[104]
NOD-SCID,
NSG, NRG
CRC1046% (NOD-SCID)
90–91% (NSG, NRG)
[105]
Ovarian cancer (high-grade serous)4376.7%[106]
NOD-SCID, NSGPDAC3590% (NSG)
40% (NOD-SCID)
[66]
NSG16 tumor types, such as bladder cancer, breast cancer, CRC, gastric cancer, glioblastoma, HCC, HNSCC, lung cancer, melanoma, ovarian cancer, PDAC, RCC, sarcoma324 PDXs [70]
Breast cancer83-[56]
CRC5054%[107]
HNSCC11545.2%[108]
Nasopharyngeal carcinoma3718.9%[109]
Testicular cancer838%[110]
Melanoma69462%[76]
Leukemia, lymphoma138 PDXs67.5% (B-ALL), 46.7% (T-ALL), 23.2% (AML)[79]
NSG, NRGBreast cancer10225% (P), 36% (M); 9% (ER+ P), 16% (ER+ M); 25% (HER2+ P), 33% (HER2+ M); 29% (TNBC P)[69]
NSG, NOGProstate cancer480%[111]
NOG10 tumor types, such as breast cancer,
CRC, lung cancer, PDAC and RCC
11653%[112]
Gastric cancer6224.2%[113]
CLL7 PDXs-[114]
ALL6093.3%[115]
NOG, NOG-IL2Metastatic melanoma2195% (lower engraftment in NOG-IL2)[81]
NOG-EXLAML2662%[116]
NSG,
NSG-SGM3
AML7782% (NSG-SGM3)
50% (NSG)
[86]
AML862.5% (NSG-SGM3)
37.5% (NSG)
[117]
NSG,
MISTRG
Favorable-risk AML968% (MISTRG)
0–20% (NSG)
[11]
MDS31Higher engraftment
in MISTRG
[17]
SRG-6, MISTRG-6MM30Higher engraftment
in MISTRG-6
[16]
Abbreviations: ACC, adrenocortical carcinoma; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CRC, colorectal cancer; ER, estrogen receptor; HCC, hepatocellular carcinoma; HER2, human epidermal growth factor receptor 2; HNSCC, head and neck squamous cell carcinoma; M, metastatic tumor; MDS, myelodysplastic syndromes; MM, multiple myeloma; NSCLC, non-small-cell lung cancer; P, primary tumor; PCa, prostate cancer; PCNA, proliferating cell nuclear antigen; PDAC, pancreatic ductal carcinoma; PD-1, programmed cell death protein 1; PDX, patient-derived xenograft; RCC, renal cell carcinoma; SCLC, small-cell lung cancer; TNBC, triple-negative breast cancer. Abbreviations of humanized mouse strains are listed in the legend of Figure 1.
Numerous studies have demonstrated that PDX models can accurately predict the response to targeted therapies and identify actionable targets in patient subgroups [99,118]. Preclinical investigations can also be conducted in parallel or sequentially in “avatar” models, when the PDX is established from the tumors of clinical trial participants [63,68,119,120]. Such co-clinical trials showed comparable response rates between PDX models (without a human immune system) and patients, including patients with breast cancer, lung cancer, pancreatic cancer and sarcoma [74,121,122,123,124]. A co-clinical trial of combined MEK and CDK4/6 inhibition in RAS mutant CRC demonstrated therapeutic efficacy, identified biomarkers of response, and revealed mechanisms of resistance in PDXs [125]. In another case, a PDX was generated from a metastatic breast cancer patient who became resistant to treatment with a phosphatidylinositol-3-kinase alpha (PI3Kα) inhibitor [126]. PI3Kα inhibitor therapy led to a loss of PTEN and resistance. By using a PDX nude mouse model, the authors showed that resistance could be avoided by simultaneous blockade of PI3Kα and PI3K p110β. In another case study, eribulin was identified to be effective in an avatar model and was successfully applied to a patient who was initially diagnosed with stage IIA TNBC but experienced metastatic recurrence in the liver [69]. Although the patient’s liver metastases and ascites regressed completely on eribulin, there was isolated progression in bone five months later. In summary, drug screening in avatar models is an innovative but time-consuming and expensive approach that can identify effective treatment options for cancer patients with recurrent disease.
Numerous studies show the benefit of using PDX models, even without a human immune system, to identify determinants of response or resistance to therapies. For example, a collection of 85 PDXs from mCRC patients was established in NOD-SCID mice and used to discover the determinants of resistance following therapy with cetuximab, an anti-epidermal growth factor receptor (EGFR) antibody [99]. The authors identified HER2 as an effective therapeutic target in cetuximab-resistant CRC. Rehman et al. used CRC PDXs and standard-of-care chemotherapy treatment to study CRC cells that become drug-tolerant persisters (DTPs) in NOD-SCID mice [127]. They showed that CRC cells possessed an equipotent capacity to enter a DTP state, and they identified therapeutic opportunities to target DTPs. Additional examples of clinically approved first- and second-generation anticancer drugs that have been successfully tested in preclinical xenograft models has been summarized by Ocana et al. and Byrne et al. [63,128].
In summary, PDX models represent a valuable resource for preclinical drug testing, biomarker validation/discovery, translational research and precision oncology [71,129]. Using PDX models to identify biomarkers of response to therapies has the potential to inform patient enrollment in clinical trials, thereby reducing the cost and time required to study responses in thousands of patients with different (sub)types of cancer.

3. Human Immune System Development and Function in Next-Generation Humanized Mice

PDX models represent a useful in vivo platform that accurately predicts the response of therapies that target tumor cells. However, PDX models do not allow study of the interaction of human tumor cells with the human immune system and assessment of the effectiveness of immune cell-targeting cancer therapies. Immunodeficient mice such as NSG, NOG, NRG, B6RGS, BRGS and SRG mice not only support the growth of PDXs, but are also capable of developing a diverse human immune system after transplantation of human CD34+ HSPCs [7,28,31,34].
Humanized PDX mouse models, which we define here as PDX mice with a human immune system, represent a promising in vivo platform for translational immuno-oncology research, testing cancer immunotherapies, and studying tumor immune escape and immune cell-driven tumor evolution and metastasis [130,131,132]. There is a large variety of humanized mouse strains available and 47 of the most prominent models are shown in Figure 1 using a genealogical tree scheme. Based on the basic immunodeficient models, next-generation humanized mice have been developed that express human cytokines, human leukocyte antigen (HLA) molecules or have other genetic modifications to support human immune cell development and function. Depending on the nature of the preclinical study, the appropriate humanized mouse model should be selected.
The development of a diverse and functional human immune system in humanized mice depends on three key principles: (1) preventing rejection of human cells (e.g., human tumor cells or human immune cells), (2) enabling of human CD34+ HSPC engraftment in the mouse bone marrow niche, and (3) supporting the development and function of human immune cell lineages [133]. An immunodeficient mouse model that lacks mouse T, B and NK cells and that provides phagocytic tolerance (e.g., via SIRPANOD, human SIRPA transgene (SIRPATg), human SIRPA knock-in (SIRPAKI) or mouse CD47−/−) is necessary to prevent rejection of transplanted human immune and tumor cells [33]. The corresponding humanized mouse models that fulfill these basic criteria are NSG [28], NOG [30], NRG [31], SRG [14,34], BRGS [134], B6RGSNOD [35], B6RGS [135] and B6RG-CD47 mice [136]. In addition, conditioning of the mouse bone marrow niche is necessary to support long-term engraftment of human CD34+ HSPCs. This is accomplished by sublethal irradiation or, less commonly, treatment with myeloablative drugs, such as busulfan [137]. Conditioning of the mouse bone marrow by sublethal irradiation or drug treatment can be avoided by genetic engineering of humanized mice. Introduction of the KitW41 [138,139] or KitWv mutation (B6RGSNOD-K [140]) or by expression of human thrombopoietin (THPO) in MISTRG mice using a knock-in (human THPO)/knock-out (mouse THPO) strategy [18] (Figure 1) renders the mouse bone marrow niche susceptible to long-term engraftment of HSPCs without the need for sublethal irradiation. However, a higher proportion of non-irradiated MISTRG humanized mice show poor human immune cell reconstitution (less than 10% of human CD45+ cells among all CD45+ cells in the blood) compared to irradiated MISTRG mice [18].
Human CD34+ cells can be obtained from umbilical cord, fetal liver, bone marrow or mobilized peripheral blood and different CD34+ HSPC engraftment procedures and kinetics have been described [130,141,142]. However, adult CD34+ cells have a 3- to 10-fold lower rate of human immune cell reconstitution compared to fetal/neonatal CD34+ cells [18,143]. The limited availability of CD34+ cells from cancer patients, together with the significantly lower immune cell reconstitution capacity of adult CD34+ cells, represents a major obstacle to the generation of PDX mice with an autologous human immune system. Human peripheral blood mononuclear cells (PBMCs) are more readily available from cancer patients than CD34+ HSPCs. However, the caveat of engrafting humanized mice with human PBMCs (hu-PBL) is the development of graft-versus-host disease (GVHD), because the transferred mature human T cells have not been educated in the mouse environment. To delay the onset of GVHD, humanized mice that lack mouse major histocompatibility complex MHC class I and/or II molecules (e.g., NSG-B2Mnull IA/IEnull, NOG-B2Mnull IAnull) [144,145] and/or that express HLAs have been generated (e.g., Rag2−/− Il2rg−/− Prf1−/− B2M−/− HLA-A2/DR1+ called “HUMAMICE”) [146]. A delayed onset of GVHD has also been observed in hu-PBL B6RG-CD47 mice, which lack mouse CD47 [147]. Another imitation of hu-PBL mouse models is the low number of human B cells and the absence of human myeloid and NK cells one week post PBMC engraftment, due to the lack of de novo generation of human immune cells (Figure 3). Improved human thymopoiesis and T cell responses have been observed in HSPC-engrafted mice that express human HLA class I and II molecules, such as NSG-A2 [148], NOG-DR4 [50], BRGS-A2/DR2 [48] and “HUMAMICE”, with a B6RG background [146] (Figure 1).
Cytokines (e.g., IL-6 and IL-7) and B cell-activating factor (BAFF) have been suggested to be important for human B cell development and/or survival in humanized mice [142]. Indeed, SRG-6 mice that express human IL-6 show not only enhanced thymopoiesis and peripheral T cell engraftment, but also increased class-switched memory B cells and serum immunoglobulin G levels [151]. The effect of human IL-6 on B cells in NSG-IL6 mice remains to be determined [152]. However, replacing mouse BAFF with human BAFF did not improve B cell maturation [153]. In 2014, complete humanization of mouse immunoglobulin loci was achieved, which enables efficient therapeutic antibody discovery [154].
IL-15 is essential for the development, maturation and function of NK cells and promotes the survival of memory CD8+ T cells, including tissue-resident CD8+ T cells [155]. Although CD56bright CD16 and CD56dim CD16+ NK cell subsets develop in NSG and SRG mice [14], functional NK cells that mediate antibody-dependent cellular cytotoxicity (ADCC), lyse HLA class I negative tumor cells or produce interferon gamma (IFN-γ) upon infection have only been observed in humanized mice that express human IL-2 (NOG-IL2 [156]), human IL-15 (SRG-15 [14], NOG-IL15 [42], NSG-IL15 [41], NSG-IL7-IL15 [40]) or in MISTRG mice, where functional IL-15-expressing human macrophages develop [18].
Myeloid cells, such as monocytes, macrophages, dendritic cells and neutrophils, are dependent on several factors for their proper development, survival and function. In particular IL-3, GM-CSF, granulocyte colony-stimulating factor (G-CSF), M-CSF and FMS-like tyrosine kinase 3 (Flt3) ligand have been shown to be important for myeloid cells, and these murine cytokines share less than 80% of their amino acid sequence compared to their human cytokine counterparts [142]. In order to improve the human myeloid compartment, mouse models that express a select combination of these human cytokines have been generated, such as MISTRG [18], NSG-SGM3 [117,149], NRG-SGM3 [157], NOG-EXL [150] and NSG-Quad mice [158]. MISTRG (SRG mice with human M-CSF, IL-3/GM-CSF and THPO) show high engraftment of HSPCs without irradiation, increased development and function of human myeloid cells and NK cells [142], and support engraftment of hematological neoplasms as mentioned in chapter two. NOG-EXL mice (NOG mice with human IL-3 and GM-CSF) show improved development of mature human basophils and mast cells, and enable the study of human allergic responses [150]. NSG-SGM3 mice (NSG mice with human SCF, IL-3 and GM-CSF) show increased human myelopoiesis and functional mast cells, but also an exhaustion of HSPCs [92,149,159]. The BRGF/BRGSF mouse model [160,161] lacks mouse Flt3 and therefore has decreased murine myelopoiesis. Exogeneous supplementation of human Flt3 ligand improved human dendritic cell development. This mouse model could thus be used to test dendritic cell-targeted therapies. In addition, MISTRG-GR [162] and NOG-GCSF mice [163] have been developed, which express human G-CSF and lack mouse G-CSF receptor (G-CSFR), and therefore support human granulocyte development and function.
In summary, a variety of next-generation humanized mouse models have been developed that should enable more authentic reconstruction of the human TIME and preclinical testing of combination (immuno)therapies. A feasible alternative to using human cytokine transgenic or knock-in mouse models is hydrodynamic injection of plasmids encoding human cytokines into immunodeficient mice [133,164].

4. Humanized PDX Mouse Models for Preclinical Testing of Cancer Immunotherapies

Currently, there are around 1500 clinical trials assessing different types of cancer immunotherapies, such as immune checkpoint inhibitors (ICIs), antibody-based therapeutics, immunomodulatory drugs and cytokines, CAR cells, therapeutic cancer vaccines and oncolytic viruses (ClinicalTrials.gov; 20 January 2023). Faithful modeling of patient tumor-immune cell interactions and responsiveness/resistance to immunotherapies in humanized PDX mouse models could thus provide critical information on the efficacy of new immunotherapeutic drugs for different cancer types and identify predictive biomarkers. With the development of a large number of new immunotherapeutics, preclinical screening of highly effective combination treatments and prevention of cross-resistance to therapies for specific cancer types will be keys to guide clinical trial design and ensure enrollment of suitable cancer patients. For example, acquired resistance to anti-MAPK therapy has been shown to confer an immune evasive tumor microenvironment and cross-resistance to immunotherapy in melanoma [165,166]. This highlights the need to test for cross-resistance between unrelated therapies and provides a rationale for treating patients with immunotherapy before they acquire resistance to anti-MAPK therapy. Preclinical testing in PDX mouse models has already led to important advancements in cancer therapy and clinical approval of immunotherapeutic agents. For example, anti-CD19 CAR-T cells successfully eradicated CD19+ B cell leukemia in PDX NSG mice [167]. The development of next-generation humanized mouse models expressing a variety of human genes that support human immune cell development and function will enable more faithful modeling of human tumor-immune cell interactions and immunotherapies that target different immune cell lineages.
Table 2. Humanized PDX mouse models used to test immunotherapeutic approaches.
Table 2. Humanized PDX mouse models used to test immunotherapeutic approaches.
Mouse ModelType of PDXType of
Immunotherapy
Human Immune System ReconstitutionReferences
NOD-SCIDNSCLCNon-autologous PBMCs + anti-PD-L1PBMCs[104]
NSGALLAnti-CD19 antibody;
anti-CD19-TRAIL fusion antibody
No[168,169]
Bladder cancer, NSCLC,
sarcoma, TNBC
Partially HLA-matched HSPCs
+ anti-PD-1 antibody
HSPCs[170]
CLLAutologous PBMCs + anti-CD38 antibodyPBMCs[171]
Dedifferentiated liposarcomaAnti-PD-1 antibodyHSPCs[172]
Gastric cancerMesothelin-specific CAR NK cellsNo[173]
HCCPartially HLA-A/B-matched HSPCs
+ anti-PD-1 or anti-CTLA-4
HSPCs[174]
HNSCCAdoptive transfer of NK cells
+ anti-PCNA antibody
No[175]
Nasopharyngeal carcinomaAnti-PD-1, anti-CTLA-4HSPCs[109]
NeuroblastomaAdoptive transfer of activated NK cells
+ anti-GD2 antibody
No[176]
Ovarian cancerAutologous TILs + anti-PD-1 antibodyTILs[177]
TNBCACT (cytokine-induced killer cells)No[178]
NRGHNSCCRadio-immunotherapy:
anti-EGFR Ab labeled with 177Lu
No[179]
BRGOsteosarcomaGD2- or HER2-targeting BiTE antibody
± anti-PD-1 or anti-PD-L1 antibody
No[180]
BRGSACC, CRC, melanoma, PDAC, SCLC, TNBCAnti-PD-1 therapy ± TKIs/WNTi/
VEGFi/HDACi
HSPCs[132]
CRCAnti-PD-1 ± cabozantinib,
anti-PD-L1 + cobimetinib
HSPCs[181]
CRCAnti-PD-1 antibodyHSPCs[182]
ACCAnti-PD-1 antibodyHSPCs[183]
NOG-IL2Metastatic melanomaACTPatient TILs[184]
NOG, NOG-IL2Metastatic melanomaACT, anti-PD-1 antibodyPatient TILs[81]
NOG-IL2Uveal melanomaAnti-HER2 CAR T cellsNo[185]
NOG-EXLAMLCo-clinical trial with BETi mivebresibNo[116]
NOG-EXL,
NSG-SGM3
Breast cancerTLR7/8 agonist activates
tumor-infiltrating pDCs
HSPCs[186]
NSG-SGM3B-ALLAnti-PD-1 +
anti-CD19 bi-specific T cell engager
HSPCs[187]
MISTRGMelanoma (CDX)Anti-VEGF antibody [18]
AMLAnti-CD117 CAR T cellsHSPCs[188]
AMLAdoptive transfer of
CBFB-MYH11-specific T cells
No[189]
HLA-deficient neuroblastomaAnti-GD2 antibodyHSPCs[190]
MISTRG-6DLBCLAnti-IL-6R antibodyHSPCs[191]
Abbreviations: Ab, antibody; ACC, adrenocortical carcinoma; ACT, adoptive cell therapy; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; B-ALL, B cell acute lymphoblastic leukemia; BETi, BET inhibitor; BiTE, bispecific T cell engager; CAR, chimeric antigen receptor; CC, cervical cancer; CDX, cell line-derived xenograft; CLL, chronic lymphocytic leukemia; CRC, colorectal cancer; CTLA-4, cytotoxic T lymphocyte-associated protein 4; DC, dendritic cell; DLBCL, diffuse large B cell lymphoma; GC, gastric cancer; GD2, disialoganglioside; GVHD, graft-versus-host disease; HDACi, histone deacetylase inhibitor; HNSCC, head and neck squamous cell carcinoma; HSPC, hematopoietic stem and progenitor cell; Ig, immunoglobulin; IL-6R, interleukin 6 receptor; mAb, monoclonal antibody; MDS, myelodysplastic syndromes; MM, multiple myeloma; NK, natural killer; NPC, nasopharyngeal carcinoma; NSCLC, non-small-cell lung cancer; OC, ovarian cancer; PBMC, peripheral blood mononuclear cell; PCa, prostate cancer; PCNA, proliferating cell nuclear antigen; PDAC, pancreatic ductal carcinoma; PD-1, programmed cell death protein 1; PD-L1, programmed cell death 1 ligand 1; pDC, plasmacytoid dendritic cells; PDX, patient-derived xenograft; RCC, renal cell carcinoma; SCLC, small-cell lung cancer; siRNA, small interfering RNA; TCR, T cell receptor; TIL, tumor-infiltrating lymphocyte; TKIs, tyrosine kinase inhibitors; TNBC, triple-negative breast cancer; VEGF, vascular endothelial growth factor; VEGFi, VEGF inhibitor; WNTi, Wnt signaling pathway inhibitor. Abbreviations of humanized mouse strains are listed in the legend of Figure 1.
In the past decade, tumor-infiltrating myeloid cells, including tumor-associated macrophages (TAMs), have emerged as critical regulators of the TIME [192,193]. They promote metastasis and therapy resistance, and M2 TAMs and polymorphonuclear myeloid-derived suppressor cells (PMN MDSCs) are associated with poor clinical prognosis in many cancer types [194,195]. TAMs can also produce immunosuppressive molecules, such as IL-10, transforming growth factor (TGF)-β, inhibitory checkpoint molecules like programmed-death ligand 1 (PD-L1) as well as angiogenic factors, such as vascular endothelial growth factor (VEGF) [196,197]. Efficient development of human myeloid cell lineages in humanized mice is therefore important to faithfully model an immunosuppressive tumor microenvironment, angiogenesis and metastasis.
Using a humanized CDX mouse model, human macrophages were shown to infiltrate melanoma CDXs in humanized MISTRG mice and promote tumor growth [18] and metastasis [19]. Increased tumor growth was dependent on human VEGF produced by tumor-infiltrating human macrophages. Treatment with the anti-human VEGF antibody bevacizumab or depletion of human phagocytic cells with clodronate reversed the increased tumor growth in MISTRG humanized mice (Table 2, Figure 4). However, the development of functional human macrophages in HSPC-engrafted MISTRG mice also caused anemia and loss of reconstitution over time, which limits the time available to perform experiments and evaluate the efficacy of therapies [18]. Human KIT+ myeloid cells were shown to facilitate metastasis in melanoma CDX NSG-SGM3 but not NSG humanized mice, which lack human factors that promote efficient human myeloid cell development [92,198]. In both, MISTRG and NSG-SGM3 humanized mouse models, the melanoma-infiltrating myeloid cells were transcriptionally similar to myeloid cells found in melanoma patients [19,198]. Using a NOD/SCID humanized mouse model, Su et al. demonstrated that CCL18 usually produced by TAMs contributes to epithelial-mesenchymal transition (EMT) and metastasis in a breast cancer CDX model [199]. However, when analyzing tumor-infiltrating plasmacytoid dendritic cells (pDCs), the intratumoral frequency of pDCs was dependent on the tumor type rather than on the humanized mouse model (NOG vs. NOG-EXL) [186]. NOG and NOG-EXL humanized mice equally supported human myeloid cell infiltration into three different human ovarian and breast cancer CDXs.
In humanized PDX mouse models, the frequency of human CD45+ cell infiltration into tumors was dependent on the individual PDX, as only 6 of 11 microsatellite stable (MSS) CRC PDXs showed an infiltration with human CD45+ cells of greater than 0.1% [132]. TNBC PDXs in NSG humanized mice showed infiltration of human T cells and CD68+ myeloid cells and generated lung metastases similar to those of TNBC patients [200]. In a study by Scherer et al., human ER+ breast cancer PDX NSG-SGM3 humanized mice could recapitulate the lymphocyte-excluded and myeloid-rich TIME of ER+ breast cancer patients [83]. Similarly, more CD33+ myeloid cells infiltrated a melanoma PDX in humanized NSG-SGM3 compared to NSG mice [201], and a neuroblastoma PDX MISTRG humanized mouse model recapitulated the lack of activated human NK cells observed in neuroblastoma patients [190]. Interestingly, even NSG mice without a human immune system partially recapitulated patient melanoma genotype-associated histopathological features [202] and allowed the analysis of metastasis-associated features of orthotopically engrafted CRC PDXs [203]. Proper development of human myeloid cells will pave the way for testing myeloid-specific phagocytosis checkpoint inhibitors. For example, blockade of the phagocytosis checkpoint SIRPA, which binds to CD47 that is upregulated on tumor cells [204], has been shown to enhance myeloid cell-dependent killing of Burkitt’s lymphoma through antibody-dependent cellular phagocytosis in SRG mice [205]. Together, these studies indicate that humanized PDX mouse models are able to recapitulate features of the patient’s TIME and may therefore be useful for preclinical testing of the efficacy of immune-based therapies and the discovery of predictive biomarkers.
Immunotherapy in humanized PDX mouse models includes anti-PD-1 therapy of bladder cancer [170], hepatocellular carcinoma (HCC) [174], melanoma [206], non-small-cell lung cancer (NSCLC) [170,207,208], autologous renal cell carcinoma (RCC) [209], sarcoma [170,172] and TNBC [200] in humanized NSG mice, as well as adrenocortical carcinoma [183], microsatellite instable (MSI) and MSS CRC [132,182], melanoma [132] and TNBC [132] in humanized BRGS mice (Table 2). Anti-PD-1 therapy in these models reduced tumor growth in approximately 75% of PDXs and led to changes in tumor-infiltrating lymphocytes and IFN-γ-related genes. In the case of TNBC PDXs, therapy responsiveness seemed to correlate with PD-L1 expression on tumor cells [200]. By using TNBC CDX BRGS humanized mice, Tentler et al. showed that RX-5902, a novel β-catenin modulator, improved the responsiveness to PD-1 or CTLA-4 inhibitor therapy [210]. Using NSG mice, HLA-A-matched CD34+ cells, fetal thymus and DNA-based vectors to provide human cytokines, Somasundaram et al. demonstrated that co-recruitment of mast cells and forkhead box P3 (FoxP3)+ regulatory T cells into melanomas was associated with resistance to anti-PD-1 therapy [211]. Combination of anti-PD-1 therapy with the tyrosine kinase inhibitors sunitinib or imatinib led to the depletion of mast cells and complete regression of tumors. By using MSI CRC patient-derived organoids orthotopically injected into NSG humanized mice, Kücükköse et al. showed that anti-PD-1 or anti-CTLA-4 therapy reduced primary CRC size and eradicated liver metastases but had no effect on peritoneal metastases [212]. This humanized PDX mouse model enabled multiorgan metastasis and suggested that high levels of immunosuppressive cytokines in ascites might promote resistance of peritoneal metastases to ICI therapy.
A major challenge for immuno-oncology in the coming years will be to identify the most effective combination immunotherapies for each cancer type and to identify treatment regimens that prevent cross-resistance between different therapies. So far, combination therapy of the CD19-directed bi-specific T cell engager blinatumomab and anti-PD-1 antibody pembrolizumab has shown improved survival in B-cell acute lymphoblastic leukemia PDX NSG-SGM3 mice [187]. Combination of anti-PD-1 antibody with histone deacetylase inhibitors has also shown successful treatment outcomes in breast cancers [182]. Cabozantinib, a multi-tyrosine kinase inhibitor, sensitized MSS CRC PDXs to anti-PD-1 therapy by increasing the frequency of cytotoxic and IFN-γ+ T cells in BRGS humanized mice [181]. This treatment also decreased the expression of T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), which is an inhibitor immune checkpoint and a marker of exhaustion on CD8+ T cells [213,214].
Another critical component of the TME are cancer-associated fibroblasts (CAFs), which can facilitate metastasis, therapeutic resistance and dormancy [215]. CAFs consist of different subpopulations with distinct phenotypes, functions and origins. Because the interaction of CAFs with tumor and immune cells critically shapes the TME and response to (immuno)therapies, it is important to properly model CAFs in humanized PDX mice. However, human CAFs that are present in the primary tumor graft are gradually replaced by mouse fibroblasts during in vivo passaging [105,216]. In a proof-of-principle study, Weinberg and colleagues used NOD-SCID mice to demonstrate that mesenchymal stromal cells (MSCs) promote breast cancer metastasis via the chemokine CCL5 [217]. Human CAFs were also shown to drive tumor progression in NSCLC-PDX NOD-SCID mice [218], and CAF subsets have been associated with (chemo)therapy resistance in TNBC and NSCLC PDX mouse models [104,219]. Of note, it may be challenging to support the long-term survival of transferred human MSCs [90], although co-injection of patient tumors and MSCs into 3D scaffolds improves the maintenance of transferred human MSCs.
Taken together, these studies demonstrate the usefulness of humanized PDX mouse models for preclinical screening of the efficacy of immunotherapies in different cancer types and subtypes.

5. Conclusions and Future Perspectives

In the past decade, numerous next-generation humanized mouse models have been generated, each with distinct improvements to enable the development of a more diverse and functional adaptive or innate immune system, or to facilitate the generation of PDXs from a wider variety of cancer types. This will ultimately accelerate preclinical screening of new immunotherapies and the discovery of biomarkers and effective treatment regimens and combination therapies. However, the unique properties (improvements and shortcomings) of each humanized mouse model must be considered in order to faithfully reconstruct the patient’s TIME and to obtain meaningful results that can be transferred to the clinic (Table 2). For example, CAR-T cell, CAR-NK cell or tumor-infiltrating lymphocytes (TILs) therapies should be performed in humanized mouse models expressing human cytokines that support T or NK cell survival and function, such as IL-2 or IL-15 [184]. In addition, adoptive T cell therapy (e.g., CAR-T cells, TILs) is routinely being performed in mice devoid of a diverse, endogenous human immune system, therefore underestimating the impact of the immunosuppressive tumor microenvironment on the function and persistence of the adoptively transferred T cells, or the ability of these T cells to infiltrate PDXs.
Despite advances in the field, some challenges and limitations remain regarding the translational value of next-generation humanized mice, such as HLA class I and II incompatibility between PDX and HSPC, and the expensive and time-consuming generation of humanized PDX mouse models. Nonetheless, the research studies highlighted in this review demonstrate the potential of humanized PDX mouse models to address key questions in precision immuno-oncology and to assist in identifying effective combination (immuno)therapies and treatment regimens for different cancer types and subtypes.

Author Contributions

Conceptualization, D.H.-B.; writing—original draft, A.C. and I.N.; writing—review and editing, D.H.-B.; funding acquisition, D.H.-B. All authors have read and agreed to the published version of the manuscript.

Funding

This research has been funded by the Austrian Science Fund (FWF) doc.funds PhD program IPPTO (DOC 59-B33; D.H.-B.) and the Vienna Science and Technology Fund (WWTF) [10.47379/LS20042] (D.H.-B.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Open Access Funding by the Austrian Science Fund (FWF). Figures created with BioRender.com.

Conflicts of Interest

D.H.-B. is co-inventor on patent application US15/097,239 (SRG-15 humanized mice). The authors declare no other conflict of interest.

References

  1. Sun, Z.; Denton, P.W.; Estes, J.D.; Othieno, F.A.; Wei, B.L.; Wege, A.K.; Melkus, M.W.; Padgett-Thomas, A.M.; Zupancic, A.; Haase, T.; et al. Intrarectal Transmission, Systemic Infection, and Cd4+ T Cell Depletion in Humanized Mice Infected with Hiv-1. J. Exp. Med. 2007, 204, 705–714. [Google Scholar] [CrossRef] [PubMed]
  2. Deng, K.; Pertea, M.; Rongvaux, A.; Wang, L.; Durand, C.M.; Ghiaur, G.; Lai, J.; McHugh, H.L.; Hao, H.; Zhang, H.; et al. Broad Ctl Response Is Required to Clear Latent Hiv-1 Due to Dominance of Escape Mutations. Nature 2015, 517, 381–385. [Google Scholar] [CrossRef] [PubMed]
  3. Sefik, E.; Israelow, B.; Mirza, H.; Zhao, J.; Qu, R.; Kaffe, E.; Song, E.; Halene, S.; Meffre, E.; Kluger, Y.; et al. A Humanized Mouse Model of Chronic COVID-19. Nat. Biotechnol. 2022, 40, 906–920. [Google Scholar] [CrossRef] [PubMed]
  4. Sefik, E.; Qu, R.; Junqueira, C.; Kaffe, E.; Mirza, H.; Zhao, J.; Brewer, J.R.; Han, A.; Steach, H.R.; Israelow, B.; et al. Inflammasome Activation in Infected Macrophages Drives Covid-19 Pathology. Nature 2022, 606, 585–593. [Google Scholar] [CrossRef]
  5. Bissig, K.-D.; Wieland, S.F.; Tran, P.; Isogawa, M.; Le, T.T.; Chisari, F.; Verma, I.M. Human Liver Chimeric Mice Provide a Model for Hepatitis B and C Virus Infection and Treatment. J. Clin. Investig. 2010, 120, 924–930. [Google Scholar] [CrossRef]
  6. Dorner, M.; Horwitz, J.A.; Donovan, B.M.; Labitt, R.N.; Budell, W.C.; Friling, T.; Vogt, A.; Catanese, M.T.; Satoh, T.; Kawai, T.; et al. Completion of the Entire Hepatitis C Virus Life Cycle in Genetically Humanized Mice. Nature 2013, 501, 237–241. [Google Scholar] [CrossRef]
  7. Traggiai, E.; Chicha, L.; Mazzucchelli, L.; Bronz, L.; Piffaretti, J.-C.; Lanzavecchia, A.; Manz, M.G. Development of a Human Adaptive Immune System in Cord Blood Cell-Transplanted Mice. Science 2004, 304, 104–107. [Google Scholar] [CrossRef]
  8. Jaiswal, S.; Pearson, T.; Friberg, H.; Shultz, L.D.; Greiner, D.L.; Rothman, A.L.; Mathew, A. Dengue Virus Infection and Virus-Specific Hla-A2 Restricted Immune Responses in Humanized Nod-Scid Il2rgammanull Mice. PLoS ONE 2009, 4, e7251. [Google Scholar] [CrossRef]
  9. Song, Y.; Shan, L.; Gbyli, R.; Liu, W.; Strowig, T.; Patel, A.; Fu, X.; Wang, X.; Xu, M.L.; Gao, Y.; et al. Combined Liver-Cytokine Humanization Comes to the Rescue of Circulating Human Red Blood Cells. Science 2021, 371, 1019–1025. [Google Scholar] [CrossRef]
  10. Barabé, F.; Kennedy, J.A.; Hope, K.J.; Dick, J.E. Modeling the Initiation and Progression of Human Acute Leukemia in Mice. Science 2007, 316, 600–604. [Google Scholar] [CrossRef]
  11. Ellegast, J.M.; Rauch, P.J.; Kovtonyuk, L.V.; Müller, R.; Wagner, U.; Saito, Y.; van Wijk, N.W.-V.; Fritz, C.; Rafiei, A.; Lysenko, V.; et al. Inv(16) and Npm1mut Amls Engraft Human Cytokine Knock-in Mice. Blood 2016, 128, 2130–2134. [Google Scholar] [CrossRef]
  12. Saito, Y.; Mochizuki, Y.; Ogahara, I.; Watanabe, T.; Hogdal, L.; Takagi, S.; Sato, K.; Kaneko, A.; Kajita, H.; Uchida, N.; et al. Overcoming Mutational Complexity in Acute Myeloid Leukemia by Inhibition of Critical Pathways. Sci. Transl. Med. 2017, 9, eaao1214. [Google Scholar] [CrossRef]
  13. Dobrovolsky, D.; Wang, E.S.; Morrow, S.; Leahy, C.; Faust, T.; Nowak, R.P.; Donovan, K.A.; Yang, G.; Li, Z.; Fischer, E.S.; et al. Bruton Tyrosine Kinase Degradation as a Therapeutic Strategy for Cancer. Blood 2019, 133, 952–961. [Google Scholar] [CrossRef] [PubMed]
  14. Herndler-Brandstetter, D.; Shan, L.; Yao, Y.; Stecher, C.; Plajer, V.; Lietzenmayer, M.; Strowig, T.; de Zoete, M.R.; Palm, N.W.; Chen, J.; et al. Humanized Mouse Model Supports Development, Function, and Tissue Residency of Human Natural Killer Cells. Proc. Natl. Acad. Sci. USA 2017, 114, E9626–E9634. [Google Scholar] [CrossRef] [PubMed]
  15. Matsui, W.; Huff, C.A.; Wang, Q.; Malehorn, M.T.; Barber, J.; Tanhehco, Y.; Smith, B.D.; Civin, C.I.; Jones, R.J. Characterization of Clonogenic Multiple Myeloma Cells. Blood 2004, 103, 2332–2336. [Google Scholar] [CrossRef]
  16. Das, R.; Strowig, T.; Verma, R.; Koduru, S.; Hafemann, A.; Hopf, S.; Kocoglu, M.H.; Borsotti, C.; Zhang, L.; Branagan, A.; et al. Microenvironment-Dependent Growth of Preneoplastic and Malignant Plasma Cells in Humanized Mice. Nat. Med. 2016, 22, 1351–1357. [Google Scholar] [CrossRef] [PubMed]
  17. Song, Y.; Rongvaux, A.; Taylor, A.; Jiang, T.; Tebaldi, T.; Balasubramanian, K.; Bagale, A.; Terzi, Y.K.; Gbyli, R.; Wang, X.; et al. A Highly Efficient and Faithful Mds Patient-Derived Xenotransplantation Model for Pre-Clinical Studies. Nat. Commun. 2019, 10, 366. [Google Scholar] [CrossRef]
  18. Rongvaux, A.; Willinger, T.; Martinek, J.; Strowig, T.; Gearty, S.; Teichmann, L.L.; Saito, Y.; Marches, F.; Halene, S.; Palucka, A.K.; et al. Development and Function of Human Innate Immune Cells in a Humanized Mouse Model. Nat. Biotechnol. 2014, 32, 364–372. [Google Scholar] [CrossRef]
  19. Voillet, V.; Berger, T.R.; McKenna, K.M.; Paulson, K.G.; Tan, W.H.; Smythe, K.S.; Hunter, D.S.; Valente, W.J.; Weaver, S.; Campbell, J.S.; et al. An In Vivo Model of Human Macrophages in Metastatic Melanoma. J. Immunol. 2022, 209, 606–620. [Google Scholar] [CrossRef]
  20. Odunsi, A.; McGray, A.J.R.; Miliotto, A.; Zhang, Y.; Wang, J.; Abiola, A.; Eppolito, C.; Huang, R.-Y. Fidelity of Human Ovarian Cancer Patient-Derived Xenografts in a Partially Humanized Mouse Model for Preclinical Testing of Immunotherapies. J. Immunother. Cancer 2020, 8, e001237. [Google Scholar] [CrossRef]
  21. Norelli, M.; Camisa, B.; Barbiera, G.; Falcone, L.; Purevdorj, A.; Genua, M.; Sanvito, F.; Ponzoni, M.; Doglioni, C.; Cristofori, P.; et al. Monocyte-Derived Il-1 and Il-6 Are Differentially Required for Cytokine-Release Syndrome and Neurotoxicity Due to Car T Cells. Nat. Med. 2018, 24, 739–748. [Google Scholar] [CrossRef]
  22. Ganesh, K.; Stadler, Z.K.; Cercek, A.; Mendelsohn, R.B.; Shia, J.; Segal, N.H.; Diaz, L.A., Jr. Immunotherapy in Colorectal Cancer: Rationale, Challenges and Potential. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 361–375. [Google Scholar] [CrossRef] [PubMed]
  23. Wong, C.H.; Siah, K.W.; Lo, A.W. Estimation of Clinical Trial Success Rates and Related Parameters. Biostatistics 2019, 20, 273–286. [Google Scholar] [CrossRef] [PubMed]
  24. De La Rochere, P.; Guil-Luna, S.; Decaudin, D.; Azar, G.; Sidhu, S.S.; Piaggio, E. Humanized Mice for the Study of Immuno-Oncology. Trends Immunol. 2018, 39, 748–763. [Google Scholar] [CrossRef] [PubMed]
  25. Flanagan, S.P. ‘Nude’, a New Hairless Gene with Pleiotropic Effects in the Mouse. Genet. Res. 1966, 8, 295–309. [Google Scholar] [CrossRef]
  26. Bosma, G.C.; Custer, R.P.; Bosma, M.J. A Severe Combined Immunodeficiency Mutation in the Mouse. Nature 1983, 301, 527–530. [Google Scholar] [CrossRef] [PubMed]
  27. Shultz, L.D.; Schweitzer, P.A.; Christianson, S.W.; Gott, B.; Schweitzer, I.B.; Tennent, B.; McKenna, S.; Mobraaten, L.; Rajan, T.V.; Greiner, D.L. Multiple Defects in Innate and Adaptive Immunologic Function in Nod/Ltsz-Scid Mice. J. Immunol. 1950, 154, 180–191. [Google Scholar] [CrossRef]
  28. Ito, M.; Hiramatsu, H.; Kobayashi, K.; Suzue, K.; Kawahata, M.; Hioki, K.; Ueyama, Y.; Koyanagi, Y.; Sugamura, K.; Tsuji, K.; et al. Nod/Scid/Gamma(C)(Null) Mouse: An Excellent Recipient Mouse Model for Engraftment of Human Cells. Blood 2002, 100, 3175–3182. [Google Scholar] [CrossRef]
  29. Shultz, L.D.; Lyons, B.L.; Burzenski, L.M.; Gott, B.; Chen, X.; Chaleff, S.; Kotb, M.; Gillies, S.D.; King, M.; Mangada, J.; et al. Human Lymphoid and Myeloid Cell Development in Nod/Ltsz-Scid Il2r Gamma Null Mice Engrafted with Mobilized Human Hemopoietic Stem Cells. J. Immunol. 2005, 174, 6477–6489. [Google Scholar] [CrossRef]
  30. Dewan, M.Z.; Terashima, K.; Taruishi, M.; Hasegawa, H.; Ito, M.; Tanaka, Y.; Mori, N.; Sata, T.; Koyanagi, Y.; Maeda, M.; et al. Rapid Tumor Formation of Human T-Cell Leukemia Virus Type 1-Infected Cell Lines in Novel Nod-Scid/Gammac(Null) Mice: Suppression by an Inhibitor against Nf-Kappab. J. Virol. 2003, 77, 5286–5294. [Google Scholar] [CrossRef]
  31. Pearson, T.; Shultz, L.D.; Miller, D.; King, M.; Laning, J.; Fodor, W.; Cuthbert, A.; Burzenski, L.; Gott, B.; Lyons, B.; et al. Non-Obese Diabetic-Recombination Activating Gene-1 (Nod-Rag1 Null) Interleukin (Il)-2 Receptor Common Gamma Chain (Il2r Gamma Null) Null Mice: A Radioresistant Model for Human Lymphohaematopoietic Engraftment. Clin. Exp. Immunol. 2008, 154, 270–284. [Google Scholar] [CrossRef] [PubMed]
  32. Oettinger, M.A.; Schatz, D.G.; Gorka, C.; Baltimore, D. RAG-1 and RAG-2, Adjacent Genes That Synergistically Activate V(D)J Recombination. Science 1990, 248, 1517–1523. [Google Scholar] [CrossRef] [PubMed]
  33. Takenaka, K.; Prasolava, T.K.; Wang, J.C.; Mortin-Toth, S.M.; Khalouei, S.; Gan, O.I.; Dick, J.E.; Danska, J.S. Polymorphism in Sirpa Modulates Engraftment of Human Hematopoietic Stem Cells. Nat. Immunol. 2007, 8, 1313–1323. [Google Scholar] [CrossRef]
  34. Strowig, T.; Rongvaux, A.; Rathinam, C.; Takizawa, H.; Borsotti, C.; Philbrick, W.; Eynon, E.E.; Manz, M.G.; Flavell, R.A. Transgenic Expression of Human Signal Regulatory Protein Alpha in Rag2-/-Gamma(C)-/- Mice Improves Engraftment of Human Hematopoietic Cells in Humanized Mice. Proc. Natl. Acad. Sci. USA 2011, 108, 13218–13223. [Google Scholar] [CrossRef]
  35. Yamauchi, T.; Takenaka, K.; Urata, S.; Shima, T.; Kikushige, Y.; Tokuyama, T.; Iwamoto, C.; Nishihara, M.; Iwasaki, H.; Miyamoto, T.; et al. Polymorphic Sirpa Is the Genetic Determinant for Nod-Based Mouse Lines to Achieve Efficient Human Cell Engraftment. Blood 2013, 121, 1316–1325. [Google Scholar] [CrossRef] [PubMed]
  36. Mian, S.A.; Anjos-Afonso, F.; Bonnet, D. Advances in Human Immune System Mouse Models for Studying Human Hematopoiesis and Cancer Immunotherapy. Front. Immunol. 2021, 11, 619236. [Google Scholar] [CrossRef]
  37. Devoy, A.; Bunton-Stasyshyn, R.K.A.; Tybulewicz, V.L.J.; Smith, A.J.H.; Fisher, E.M.C. Genomically Humanized Mice: Technologies and Promises. Nat. Rev. Genet. 2011, 13, 14–20. [Google Scholar] [CrossRef]
  38. Zhu, F.; Nair, R.R.; Fisher, E.M.C.; Cunningham, T.J. Humanising the Mouse Genome Piece by Piece. Nat. Commun. 2019, 10, 1845. [Google Scholar] [CrossRef]
  39. Coppin, E.; Sundarasetty, B.S.; Rahmig, S.; Blume, J.; Verheyden, N.A.; Bahlmann, F.; Ravens, S.; Schubert, U.; Schmid, J.; Ludwig, S.; et al. Enhanced Differentiation of Functional Human T Cells in Nsgw41 Mice with Tissue-Specific Expression of Human Interleukin-7. Leukemia 2021, 35, 3561–3567. [Google Scholar] [CrossRef]
  40. Matsuda, M.; Ono, R.; Iyoda, T.; Endo, T.; Iwasaki, M.; Tomizawa-Murasawa, M.; Saito, Y.; Kaneko, A.; Shimizu, K.; Yamada, D.; et al. Human Nk Cell Development in Hil-7 and Hil-15 Knockin Nod/Scid/Il2rgko Mice. Life Sci. Alliance 2019, 2, e201800195. [Google Scholar] [CrossRef]
  41. Aryee, K.E.; Burzenski, L.M.; Yao, L.C.; Keck, J.G.; Greiner, D.L.; Shultz, L.D.; Brehm, M.A. Enhanced Development of Functional Human Nk Cells in Nod-Scid-Il2rg(Null) Mice Expressing Human Il15. FASEB J. 2022, 36, e22476. [Google Scholar] [CrossRef] [PubMed]
  42. Katano, I.; Nishime, C.; Ito, R.; Kamisako, T.; Mizusawa, T.; Ka, Y.; Ogura, T.; Suemizu, H.; Kawakami, Y.; Ito, M.; et al. Long-Term Maintenance of Peripheral Blood Derived Human Nk Cells in a Novel Human Il-15- Transgenic Nog Mouse. Sci. Rep. 2017, 7, 17230. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Y.; Masse-Ranson, G.; Garcia, Z.; Bruel, T.; Kök, A.; Strick-Marchand, H.; Jouvion, G.; Serafini, N.; Lim, A.I.; Dusseaux, M.; et al. A Human Immune System Mouse Model with Robust Lymph Node Development. Nat. Methods 2018, 15, 623–630. [Google Scholar] [CrossRef] [PubMed]
  44. Shultz, L.D.; Saito, Y.; Najima, Y.; Tanaka, S.; Ochi, T.; Tomizawa, M.; Ishikawa, F. Generation of Functional Human T-Cell Subsets with Hla-Restricted Immune Responses in Hla Class I Expressing Nod/Scid/Il2r Gamma(Null) Humanized Mice. Proc. Natl. Acad. Sci. USA 2010, 107, 13022–13027. [Google Scholar] [CrossRef]
  45. Mori, A.; Murata, S.; Tashiro, N.; Tadokoro, T.; Okamoto, S.; Otsuka, R.; Wada, H.; Murata, T.; Takahashi, T.; Seino, K.-I.; et al. Establishment of Human Leukocyte Antigen-Mismatched Immune Responses after Transplantation of Human Liver Bud in Humanized Mouse Models. Cells 2021, 10, 476. [Google Scholar] [CrossRef]
  46. Majji, S.; Wijayalath, W.; Shashikumar, S.; Pow-Sang, L.; Villasante, E.; Brumeanu, T.D.; Casares, S. Differential Effect of Hla Class-I Versus Class-Ii Transgenes on Human T and B Cell Reconstitution and Function in Nrg Mice. Sci. Rep. 2016, 6, 28093. [Google Scholar] [CrossRef]
  47. Labarthe, L.; Henriquez, S.; Lambotte, O.; Di Santo, J.P.; Le Grand, R.; Pflumio, F.; Arcangeli, M.-L.; Legrand, N.; Bourgeois, C. Frontline Science: Exhaustion and Senescence Marker Profiles on Human T Cells in Brgsf-A2 Humanized Mice Resemble Those in Human Samples. J. Leukoc. Biol. 2020, 107, 27–42. [Google Scholar] [CrossRef]
  48. Masse-Ranson, G.; Dusséaux, M.; Fiquet, O.; Darche, S.; Boussand, M.; Li, Y.; Lopez-Lastra, S.; Legrand, N.; Corcuff, E.; Toubert, A.; et al. Accelerated Thymopoiesis and Improved T-Cell Responses in Hla-A2/-Dr2 Transgenic Brgs-Based Human Immune System Mice. Eur. J. Immunol. 2019, 49, 954–965. [Google Scholar] [CrossRef]
  49. Billerbeck, E.; Horwitz, J.A.; Labitt, R.N.; Donovan, B.M.; Vega, K.; Budell, W.C.; Koo, G.C.; Rice, C.M.; Ploss, A. Characterization of Human Antiviral Adaptive Immune Responses during Hepatotropic Virus Infection in HLA-Transgenic Human Immune System Mice. J. Immunol. 2013, 191, 1753–1764. [Google Scholar] [CrossRef]
  50. Suzuki, M.; Takahashi, T.; Katano, I.; Ito, R.; Ito, M.; Harigae, H.; Ishii, N.; Sugamura, K. Induction of Human Humoral Immune Responses in a Novel Hla-Dr-Expressing Transgenic Nod/Shi-Scid/Gammacnull Mouse. Int. Immunol. 2012, 24, 243–252. [Google Scholar] [CrossRef]
  51. Serr, I.; Fürst, R.W.; Achenbach, P.; Scherm, M.G.; Gökmen, F.; Haupt, F.; Sedlmeier, E.-M.; Knopff, A.; Shultz, L.; Willis, R.A.; et al. Type 1 Diabetes Vaccine Candidates Promote Human Foxp3(+)Treg Induction in Humanized Mice. Nat. Commun. 2016, 7, 10991. [Google Scholar] [CrossRef]
  52. Johnson, J.I.; Decker, S.; Zaharevitz, D.; Rubinstein, L.V.; Venditti, J.M.; Schepartz, S.; Kalyandrug, S.; Christian, M.; Arbuck, S.; Hollingshead, M.; et al. Relationships between Drug Activity in Nci Preclinical in Vitro and in Vivo Models and Early Clinical Trials. Br. J. Cancer 2001, 84, 1424–1431. [Google Scholar] [CrossRef]
  53. Gillet, J.-P.; Calcagno, A.M.; Varma, S.; Marino, M.; Green, L.J.; Vora, M.I.; Patel, C.; Orina, J.N.; Eliseeva, T.A.; Singal, V.; et al. Redefining the Relevance of Established Cancer Cell Lines to the Study of Mechanisms of Clinical Anti-Cancer Drug Resistance. Proc. Natl. Acad. Sci. USA 2011, 108, 18708–18713. [Google Scholar] [CrossRef] [PubMed]
  54. Dangles-Marie, V.; Pocard, M.; Richon, S.; Weiswald, L.-B.; Assayag, F.; Saulnier, P.; Judde, J.-G.; Janneau, J.-L.; Auger, N.; Validire, P.; et al. Establishment of Human Colon Cancer Cell Lines from Fresh Tumors versus Xenografts: Comparison of Success Rate and Cell Line Features. Cancer Res. 2007, 67, 398–407. [Google Scholar] [CrossRef] [PubMed]
  55. Rückert, F.; Aust, D.; Böhme, I.; Werner, K.; Brandt, A.; Diamandis, E.P.; Krautz, C.; Hering, S.; Saeger, H.-D.; Grützmann, R.; et al. Five Primary Human Pancreatic Adenocarcinoma Cell Lines Established by the Outgrowth Method. J. Surg. Res. 2012, 172, 29–39. [Google Scholar] [CrossRef] [PubMed]
  56. Bruna, A.; Rueda, O.M.; Greenwood, W.; Batra, A.S.; Callari, M.; Batra, R.N.; Pogrebniak, K.; Sandoval, J.; Cassidy, J.W.; Tufegdzic-Vidakovic, A.; et al. A Biobank of Breast Cancer Explants with Preserved Intra-tumor Heterogeneity to Screen Anticancer Compounds. Cell 2016, 167, 260–274.e22. [Google Scholar] [CrossRef]
  57. Qiu, Z.; Li, H.; Zhang, Z.; Zhu, Z.; He, S.; Wang, X.; Wang, P.; Qin, J.; Zhuang, L.; Wang, W.; et al. A Pharmacogenomic Landscape in Human Liver Cancers. Cancer Cell 2019, 36, 179–193.e11. [Google Scholar] [CrossRef]
  58. Weeber, F.; Ooft, S.N.; Dijkstra, K.K.; Voest, E.E. Tumor Organoids as a Pre-clinical Cancer Model for Drug Discovery. Cell Chem. Biol. 2017, 24, 1092–1100. [Google Scholar] [CrossRef]
  59. Marusyk, A.; Janiszewska, M.; Polyak, K. Intratumor Heterogeneity: The Rosetta Stone of Therapy Resistance. Cancer Cell 2020, 37, 471–484. [Google Scholar] [CrossRef]
  60. Pearson, A.T.; Finkel, K.A.; Warner, K.A.; Nör, F.; Tice, D.; Martins, M.D.; Jackson, T.L.; Nör, J.E. Patient-Derived Xenograft (Pdx) Tumors Increase Growth Rate with Time. Oncotarget 2016, 7, 7993–8005. [Google Scholar] [CrossRef]
  61. Julien, S.; Merino-Trigo, A.; Lacroix, L.; Pocard, M.; Goéré, D.; Mariani, P.; Landron, S.; Bigot, L.; Nemati, F.; Dartigues, P.; et al. Characterization of a Large Panel of Patient-Derived Tumor Xenografts Representing the Clinical Heterogeneity of Human Colorectal Cancer. Clin. Cancer Res. 2012, 18, 5314–5328. [Google Scholar] [CrossRef] [PubMed]
  62. Zhu, M.; Li, L.; Lu, T.; Yoo, H.; Zhu, J.; Gopal, P.; Wang, S.C.; Porembka, M.R.; Rich, N.E.; Kagan, S.; et al. Uncovering Biological Factors That Regulate Hepatocellular Carcinoma Growth Using Patient-Derived Xenograft Assays. Hepatology 2020, 72, 1085–1101. [Google Scholar] [CrossRef] [PubMed]
  63. Byrne, A.T.; Alférez, D.G.; Amant, F.; Annibali, D.; Arribas, J.; Biankin, A.V.; Bruna, A.; Budinská, E.; Caldas, C.; Chang, D.K.; et al. Interrogating Open Issues in Cancer Precision Medicine with Patient-Derived Xenografts. Nat. Rev. Cancer 2017, 17, 254–268. [Google Scholar] [CrossRef] [PubMed]
  64. Ye, W.; Jiang, Z.; Li, G.-X.; Xiao, Y.; Lin, S.; Lai, Y.; Wang, S.; Li, B.; Jia, B.; Li, Y.; et al. Quantitative Evaluation of the Immunodeficiency of a Mouse Strain by Tumor Engraftments. J. Hematol. Oncol. 2015, 8, 59. [Google Scholar] [CrossRef] [PubMed]
  65. Okada, S.; Vaeteewoottacharn, K.; Kariya, R. Application of Highly Immunocompromised Mice for the Establishment of Patient-Derived Xenograft (Pdx) Models. Cells 2019, 8, 889. [Google Scholar] [CrossRef] [PubMed]
  66. Magouliotis, D.E.; Lafazanis, K.; Koutsougianni, F.; Sakellaridis, N.; Ioannou, M.; Zacharoulis, D.; Dimas, K. Establishment of Patient-derived Orthotopic Xenografts (PDX) as Models for Pancreatic Ductal Adenocarcinoma. In Vivo 2022, 36, 1114–1119. [Google Scholar] [CrossRef]
  67. Hidalgo, M.; Amant, F.; Biankin, A.V.; Budinská, E.; Byrne, A.T.; Caldas, C.; Clarke, R.B.; de Jong, S.; Jonkers, J.; Mælandsmo, G.M.; et al. Patient-Derived Xenograft Models: An Emerging Platform for Translational Cancer Research. Cancer Discov. 2014, 4, 998–1013. [Google Scholar] [CrossRef]
  68. Koga, Y.; Ochiai, A. Systematic Review of Patient-Derived Xenograft Models for Preclinical Studies of Anti-Cancer Drugs in Solid Tumors. Cells 2019, 8, 418. [Google Scholar] [CrossRef]
  69. Guillen, K.P.; Fujita, M.; Butterfield, A.J.; Scherer, S.D.; Bailey, M.H.; Chu, Z.; DeRose, Y.S.; Zhao, L.; Cortes-Sanchez, E.; Yang, C.-H.; et al. A Human Breast Cancer-Derived Xenograft and Organoid Platform for Drug Discovery and Precision Oncology. Nat. Cancer 2022, 3, 232–250. [Google Scholar] [CrossRef]
  70. Woo, X.Y.; Giordano, J.; Srivastava, A.; Zhao, Z.M.; Lloyd, M.W.; de Bruijn, R.; Suh, Y.S.; Patidar, R.; Chen, L.; Scherer, S.; et al. Conservation of Copy Number Profiles During Engraftment and Passaging of Patient-Derived Cancer Xenografts. Nat. Genet. 2021, 53, 86–99. [Google Scholar] [CrossRef]
  71. Zanella, E.R.; Grassi, E.; Trusolino, L. Towards Precision Oncology with Patient-Derived Xenografts. Nat. Rev. Clin. Oncol. 2022, 19, 719–732. [Google Scholar] [CrossRef] [PubMed]
  72. Abdirahman, S.M.; Christie, M.; Preaudet, A.; Burstroem, M.C.U.; Mouradov, D.; Lee, B.; Sieber, O.M.; Putoczki, T.L. A Biobank of Colorectal Cancer Patient-Derived Xenografts. Cancers 2020, 12, 2340. [Google Scholar] [CrossRef]
  73. Gao, H.; Korn, J.M.; Ferretti, S.; Monahan, J.E.; Wang, Y.; Singh, M.; Zhang, C.; Schnell, C.; Yang, G.; Zhang, Y.; et al. High-Throughput Screening Using Patient-Derived Tumor Xenografts to Predict Clinical Trial Drug Response. Nat. Med. 2015, 21, 1318–1325. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, X.; Claerhout, S.; Prat, A.; Dobrolecki, L.E.; Petrovic, I.; Lai, Q.; Landis, M.D.; Wiechmann, L.; Schiff, R.; Giuliano, M.; et al. A Renewable Tissue Resource of Phenotypically Stable, Biologically and Ethnically Diverse, Patient-Derived Human Breast Cancer Xenograft Models. Cancer Res. 2013, 73, 4885–4897. [Google Scholar] [CrossRef] [PubMed]
  75. Savage, P.; Pacis, A.; Kuasne, H.; Liu, L.; Lai, D.; Wan, A.; Dankner, M.; Martinez, C.; Muñoz-Ramos, V.; Pilon, V.; et al. Chemogenomic Profiling of Breast Cancer Patient-Derived Xenografts Reveals Targetable Vulnerabilities for Difficult-to-Treat Tumors. Commun. Biol. 2020, 3, 310. [Google Scholar] [CrossRef]
  76. Krepler, C.; Sproesser, K.; Brafford, P.; Beqiri, M.; Garman, B.; Xiao, M.; Shannan, B.; Watters, A.; Perego, M.; Zhang, G.; et al. A Comprehensive Patient-Derived Xenograft Collection Representing the Heterogeneity of Melanoma. Cell Rep. 2017, 21, 1953–1967. [Google Scholar] [CrossRef]
  77. Caeser, R.; Egger, J.V.; Chavan, S.; Socci, N.D.; Jones, C.B.; Kombak, F.E.; Asher, M.; Roehrl, M.H.; Shah, N.S.; Allaj, V.; et al. Genomic and Transcriptomic Analysis of a Library of Small Cell Lung Cancer Patient-Derived Xenografts. Nat. Commun. 2022, 13, 2144. [Google Scholar] [CrossRef]
  78. Risbridger, G.P.; Clark, A.K.; Porter, L.H.; Toivanen, R.; Bakshi, A.; Lister, N.L.; Pook, D.; Pezaro, C.J.; Sandhu, S.; Keerthikumar, S.; et al. The Mural Collection of Prostate Cancer Patient-Derived Xenografts Enables Discovery through Preclinical Models of Uro-Oncology. Nat. Commun. 2021, 12, 5049. [Google Scholar] [CrossRef]
  79. Townsend, E.C.; Murakami, M.A.; Christodoulou, A.; Christie, A.L.; Koster, J.; DeSouza, T.A.; Morgan, E.A.; Kallgren, S.P.; Liu, H.; Wu, S.C.; et al. The Public Repository of Xenografts Enables Discovery and Randomized Phase Ii-Like Trials in Mice. Cancer Cell 2016, 30, 183. [Google Scholar] [CrossRef]
  80. Wang, Y.; Revelo, M.P.; Sudilovsky, D.; Cao, M.; Chen, W.G.; Goetz, L.; Xue, H.; Sadar, M.; Shappell, S.B.; Cunha, G.R.; et al. Development and Characterization of Efficient Xenograft Models for Benign and Malignant Human Prostate Tissue. Prostate 2005, 64, 149–159. [Google Scholar] [CrossRef]
  81. Ny, L.; Rizzo, L.Y.; Belgrano, V.; Karlsson, J.; Jespersen, H.; Carstam, L.; Bagge, R.O.; Nilsson, L.M.; Nilsson, J.A. Supporting Clinical Decision Making in Advanced Melanoma by Preclinical Testing in Personalized Immune-Humanized Xenograft Mouse Models. Ann. Oncol. 2020, 31, 266–273. [Google Scholar] [CrossRef] [PubMed]
  82. DeRose, Y.S.; Wang, G.; Lin, Y.C.; Bernard, P.S.; Buys, S.S.; Ebbert, M.T.; Factor, R.; Matsen, C.; Milash, B.A.; Nelson, E.; et al. Tumor Grafts Derived from Women with Breast Cancer Authentically Reflect Tumor Pathology, Growth, Metastasis and Disease Outcomes. Nat. Med. 2011, 17, 1514–1520. [Google Scholar] [CrossRef]
  83. Scherer, S.D.; Riggio, A.I.; Haroun, F.; DeRose, Y.S.; Ekiz, H.A.; Fujita, M.; Toner, J.; Zhao, L.; Li, Z.; Oesterreich, S.; et al. An Immune-Humanized Patient-Derived Xenograft Model of Estrogen-Independent, Hormone Receptor Positive Metastatic Breast Cancer. Breast Cancer Res. 2021, 23, 100. [Google Scholar] [CrossRef] [PubMed]
  84. Petrosyan, V.; Dobrolecki, L.E.; LaPlante, E.L.; Srinivasan, R.R.; Bailey, M.H.; Welm, A.L.; Welm, B.E.; Lewis, M.T.; Milosavljevic, A. Immunologically "Cold" Triple Negative Breast Cancers Engraft at a Higher Rate in Patient Derived Xenografts. npj Breast Cancer 2022, 8, 104. [Google Scholar] [CrossRef] [PubMed]
  85. de la Guardia, R.D.; Velasco-Hernandez, T.; Gutiérrez-Agüera, F.; Roca-Ho, H.; Molina, O.; Nombela-Arrieta, C.; Bataller, A.; Fuster, J.L.; Anguita, E.; Vives, S.; et al. Engraftment Characterization of Risk-Stratified Aml in Nsgs Mice. Blood Adv. 2021, 5, 4842–4854. [Google Scholar] [CrossRef]
  86. Krevvata, M.; Shan, X.; Zhou, C.; Dos Santos, C.; Ndikuyeze, G.H.; Secreto, A.; Glover, J.; Trotman, W.; Brake-Silla, G.; Nunez-Cruz, S.; et al. Cytokines Increase Engraftment of Human Acute Myeloid Leukemia Cells in Immunocompromised Mice but Not Engraftment of Human Myelodysplastic Syndrome Cells. Haematologica 2018, 103, 959–971. [Google Scholar] [CrossRef]
  87. Shlush, L.I.; Mitchell, A.; Heisler, L.; Abelson, S.; Ng, S.W.K.; Trotman-Grant, A.; Medeiros, J.J.F.; Rao-Bhatia, A.; Jaciw-Zurakowsky, I.; Marke, R.; et al. Tracing the Origins of Relapse in Acute Myeloid Leukaemia to Stem Cells. Nature 2017, 547, 104–108. [Google Scholar] [CrossRef]
  88. Reinisch, A.; Thomas, D.; Corces, M.R.; Zhang, X.; Gratzinger, D.; Hong, W.-J.; Schallmoser, K.; Strunk, D.; Majeti, R. A Humanized Bone Marrow Ossicle Xenotransplantation Model Enables Improved Engraftment of Healthy and Leukemic Human Hematopoietic Cells. Nat. Med. 2016, 22, 812–821. [Google Scholar] [CrossRef]
  89. Medyouf, H.; Mossner, M.; Jann, J.-C.; Nolte, F.; Raffel, S.; Herrmann, C.; Lier, A.; Eisen, C.; Nowak, V.; Zens, B.; et al. Myelodysplastic Cells in Patients Reprogram Mesenchymal Stromal Cells to Establish a Transplantable Stem Cell Niche Disease Unit. Cell Stem Cell 2014, 14, 824–837. [Google Scholar] [CrossRef]
  90. Rouault-Pierre, K.; Mian, S.A.; Goulard, M.; Abarrategi, A.; Di Tulio, A.; Smith, A.E.; Mohamedali, A.; Best, S.; Nloga, A.-M.; Kulasekararaj, A.G.; et al. Preclinical Modeling of Myelodysplastic Syndromes. Leukemia 2017, 31, 2702–2708. [Google Scholar] [CrossRef]
  91. Nicolini, F.E.; Cashman, J.D.; Hogge, D.E.; Humphries, R.K.; Eaves, C.J. Nod/Scid Mice Engineered to Express Human Il-3, Gm-Csf and Steel Factor Constitutively Mobilize Engrafted Human Progenitors and Compromise Human Stem Cell Regeneration. Leukemia 2004, 18, 341–347. [Google Scholar] [CrossRef] [PubMed]
  92. Billerbeck, E.; Barry, W.T.; Mu, K.; Dorner, M.; Rice, C.M.; Ploss, A. Development of Human Cd4+Foxp3+ Regulatory T Cells in Human Stem Cell Factor-, Granulocyte-Macrophage Colony-Stimulating Factor-, and Interleukin-3-Expressing Nod-Scid Il2rgamma(Null) Humanized Mice. Blood 2011, 117, 3076–3086. [Google Scholar] [CrossRef] [PubMed]
  93. Svoboda, D.S.; Barrasa, M.I.; Shu, J.; Rietjens, R.; Zhang, S.; Mitalipova, M.; Berube, P.; Fu, D.; Shultz, L.D.; Bell, G.W.; et al. Human Ipsc-Derived Microglia Assume a Primary Microglia-Like State after Transplantation into the Neonatal Mouse Brain. Proc. Natl. Acad. Sci. USA 2019, 116, 25293–25303. [Google Scholar] [CrossRef] [PubMed]
  94. Sandén, C.; Lilljebjörn, H.; Pietras, C.O.; Henningsson, R.; Saba, K.H.; Landberg, N.; Thorsson, H.; von Palffy, S.; Peña-Martinez, P.; Högberg, C.; et al. Clonal Competition within Complex Evolutionary Hierarchies Shapes Aml over Time. Nat. Commun. 2020, 11, 579. [Google Scholar]
  95. Eirew, P.; Steif, A.; Khattra, J.; Ha, G.; Yap, D.; Farahani, H.; Gelmon, K.; Chia, S.; Mar, C.; Wan, A.; et al. Dynamics of Genomic Clones in Breast Cancer Patient Xenografts at Single-Cell Resolution. Nature 2015, 518, 422–426. [Google Scholar] [CrossRef]
  96. Linnebacher, M.; Maletzki, C.; Ostwald, C.; Klier, U.; Krohn, M.; Klar, E.; Prall, F. Cryopreservation of Human Colorectal Carcinomas Prior to Xenografting. BMC Cancer 2010, 10, 362. [Google Scholar] [CrossRef]
  97. Nemati, F.; Sastre-Garau, X.; Laurent, C.; Couturier, J.; Mariani, P.; Desjardins, L.; Piperno-Neumann, S.; Lantz, O.; Asselain, B.; Plancher, C.; et al. Establishment and Characterization of a Panel of Human Uveal Melanoma Xenografts Derived from Primary and/or Metastatic Tumors. Clin. Cancer Res. 2010, 16, 2352–2362. [Google Scholar] [CrossRef]
  98. Zou, S.; Ye, M.; Zhang, J.-A.; Ji, H.; Chen, Y.; Zhu, X. Establishment and Genetically Characterization of Patient-Derived Xenograft Models of Cervical Cancer. BMC Med. Genom. 2022, 15, 191. [Google Scholar] [CrossRef]
  99. Bertotti, A.; Migliardi, G.; Galimi, F.; Sassi, F.; Torti, D.; Isella, C.; Cora, D.; Di Nicolantonio, F.; Buscarino, M.; Petti, C.; et al. A Molecularly Annotated Platform of Patient-Derived Xenografts (“Xenopatients”) Identifies Her2 as an Effective Therapeutic Target in Cetuximab-Resistant Colorectal Cancer. Cancer Discov. 2011, 1, 508–523. [Google Scholar] [CrossRef]
  100. Zou, J.; Liu, Y.; Wang, J.; Liu, Z.; Lu, Z.; Chen, Z.; Li, Z.; Dong, B.; Huang, W.; Li, Y.; et al. Establishment and Genomic Characterizations of Patient-Derived Esophageal Squamous Cell Carcinoma Xenograft Models Using Biopsies for Treatment Optimization. J. Transl. Med. 2018, 16, 15. [Google Scholar] [CrossRef]
  101. Zhu, Y.; Tian, T.; Li, Z.; Tang, Z.; Wang, L.; Wu, J.; Li, Y.; Dong, B.; Li, Y.; Li, N.; et al. Establishment and Characterization of Patient-Derived Tumor Xenograft Using Gastroscopic Biopsies in Gastric Cancer. Sci. Rep. 2015, 5, 8542. [Google Scholar] [CrossRef] [PubMed]
  102. Agnusdei, V.; Minuzzo, S.A.; Frasson, C.; Grassi, A.; Axelrod, F.; Satyal, S.; Gurney, A.; Hoey, T.; Seganfreddo, E.; Basso, G.; et al. Therapeutic Antibody Targeting of Notch1 in T-Acute Lymphoblastic Leukemia Xenografts. Leukemia 2014, 28, 278–288. [Google Scholar] [CrossRef] [PubMed]
  103. Xu, C.; Yan, L.; An, Q.; Zhang, S.; Guan, X.; Wang, Z.; Lv, A.; Liu, D.; Liu, F.; Dong, B.; et al. Establishment and Evaluation of Retroperitoneal Liposarcoma Patient-Derived Xenograft Models: An Ideal Model for Preclinical Study. Int. J. Med. Sci. 2022, 19, 1241–1253. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, X.; Shen, C.; Wei, Z.; Zhang, R.; Wang, Y.; Jiang, L.; Chen, K.; Qiu, S.; Zhang, Y.; Zhang, T.; et al. Patient-Derived Non-Small Cell Lung Cancer Xenograft Mirrors Complex Tumor Heterogeneity. Cancer Biol. Med. 2021, 18, 184–198. [Google Scholar] [CrossRef]
  105. Maykel, J.; Liu, J.H.; Li, H.; Shultz, L.D.; Greiner, D.L.; Houghton, J. Nod-Scidil2rg (Tm1wjl) and Nod-Rag1 (Null) Il2rg (Tm1wjl): A Model for Stromal Cell-Tumor Cell Interaction for Human Colon Cancer. Dig. Dis. Sci. 2014, 59, 1169–1179. [Google Scholar] [CrossRef] [PubMed]
  106. Cybula, M.; Wang, L.; Wang, L.; Drumond-Bock, A.L.; Moxley, K.M.; Benbrook, D.M.; Gunderson-Jackson, C.; Ruiz-Echevarria, M.J.; Bhattacharya, R.; Mukherjee, P.; et al. Patient-Derived Xenografts of High-Grade Serous Ovarian Cancer Subtype as a Powerful Tool in Pre-Clinical Research. Cancers 2021, 13, 628. [Google Scholar] [CrossRef] [PubMed]
  107. Chou, J.; FitzGibbon, M.P.; Mortales, C.-L.L.; Towlerton, A.M.H.; Upton, M.P.; Yeung, R.S.; McIntosh, M.W.; Warren, E.H. Phenotypic and Transcriptional Fidelity of Patient-Derived Colon Cancer Xenografts in Immune-Deficient Mice. PLoS ONE 2013, 8, e79874. [Google Scholar] [CrossRef]
  108. Klinghammer, K.; Raguse, J.-D.; Plath, T.; Albers, A.E.; Joehrens, K.; Zakarneh, A.; Brzezicha, B.; Wulf-Goldenberg, A.; Keilholz, U.; Hoffmann, J.; et al. A Comprehensively Characterized Large Panel of Head and Neck Cancer Patient-Derived Xenografts Identifies the Mtor Inhibitor Everolimus as Potential New Treatment Option. Int. J. Cancer 2015, 136, 2940–2948. [Google Scholar] [CrossRef]
  109. Liu, W.N.; Fong, S.Y.; Tan, W.W.S.; Tan, S.Y.; Liu, M.; Cheng, J.Y.; Lim, S.; Suteja, L.; Huang, E.K.; Chan, J.K.Y.; et al. Establishment and Characterization of Humanized Mouse Npc-Pdx Model for Testing Immunotherapy. Cancers 2020, 12, 1025. [Google Scholar] [CrossRef]
  110. de Vries, G.; Rosas-Plaza, X.; Meersma, G.J.; Leeuwenburgh, V.C.; Kok, K.; Suurmeijer, A.J.H.; van Vugt, M.A.T.M.; Gietema, J.A.; de Jong, S. Establishment and Characterisation of Testicular Cancer Patient-Derived Xenograft Models for Preclinical Evaluation of Novel Therapeutic Strategies. Sci. Rep. 2020, 10, 18938. [Google Scholar] [CrossRef]
  111. Wetterauer, C.; Vlajnic, T.; Schuler, J.; Gsponer, J.R.; Thalmann, G.N.; Cecchini, M.; Schneider, J.; Zellweger, T.; Pueschel, H.; Bachmann, A.; et al. Early Development of Human Lymphomas in a Prostate Cancer Xenograft Program Using Triple Knock-out Immunocompromised Mice. Prostate 2015, 75, 585–592. [Google Scholar] [CrossRef] [PubMed]
  112. Chijiwa, T.; Kawai, K.; Noguchi, A.; Sato, H.; Hayashi, A.; Cho, H.; Shiozawa, M.; Kishida, T.; Morinaga, S.; Yokose, T.; et al. Establishment of Patient-Derived Cancer Xenografts in Immunodeficient Nog Mice. Int. J. Oncol. 2015, 47, 61–70. [Google Scholar] [CrossRef] [PubMed]
  113. Choi, Y.Y.; Lee, J.E.; Kim, H.; Sim, M.H.; Kim, K.-K.; Lee, G.; Kim, H.I.; An, J.Y.; Hyung, W.J.; Kim, C.-B.; et al. Establishment and Characterisation of Patient-Derived Xenografts as Paraclinical Models for Gastric Cancer. Sci. Rep. 2016, 6, 22172. [Google Scholar] [CrossRef]
  114. Davies, N.J.; Kwok, M.; Gould, C.; Oldreive, C.E.; Mao, J.; Parry, H.; Smith, E.; Agathanggelou, A.; Pratt, G.; Taylor, A.M.R.; et al. Dynamic Changes in Clonal Cytogenetic Architecture During Progression of Chronic Lymphocytic Leukemia in Patients and Patient-Derived Murine Xenografts. Oncotarget 2017, 8, 44749–44760. [Google Scholar] [CrossRef]
  115. Tanaka, K.; Kato, I.; Dobashi, Y.; Imai, J.; Mikami, T.; Kubota, H.; Ueno, H.; Ito, M.; Ogawa, S.; Nakahata, T.; et al. The First Japanese Biobank of Patient-Derived Pediatric Acute Lymphoblastic Leukemia Xenograft Models. Cancer Sci. 2022, 113, 3814–3825. [Google Scholar] [CrossRef] [PubMed]
  116. Albert, D.H.; Goodwin, N.C.; Davies, A.M.; Rowe, J.; Feuer, G.; Boyiadzis, M.; Dorritie, K.A.; Mancini, M.; Gandour-Edwards, R.; Jonas, B.A.; et al. Co-Clinical Modeling of the Activity of the Bet Inhibitor Mivebresib (Abbv-075) in Aml. In Vivo 2022, 36, 1615–1627. [Google Scholar] [CrossRef]
  117. Wunderlich, M.; Chou, F.-S.; Link, K.A.; Mizukawa, B.; Perry, R.L.; Carroll, M.; Mulloy, J.C. Aml Xenograft Efficiency Is Significantly Improved in Nod/Scid-Il2rg Mice Constitutively Expressing Human Scf, Gm-Csf and Il-3. Leukemia 2010, 24, 1785–1788. [Google Scholar] [CrossRef]
  118. Zanella, E.R.; Galimi, F.; Sassi, F.; Migliardi, G.; Cottino, F.; Leto, S.M.; Lupo, B.; Erriquez, J.; Isella, C.; Comoglio, P.M.; et al. Igf2 Is an Actionable Target That Identifies a Distinct Subpopulation of Colorectal Cancer Patients with Marginal Response to Anti-Egfr Therapies. Sci. Transl. Med. 2015, 7, 272ra12. [Google Scholar] [CrossRef]
  119. Yoshida, G.J. Applications of Patient-Derived Tumor Xenograft Models and Tumor Organoids. J. Hematol. Oncol. 2020, 13, 4. [Google Scholar] [CrossRef]
  120. Genta, S.; Coburn, B.; Cescon, D.W.; Spreafico, A. Patient-Derived Cancer Models: Valuable Platforms for Anticancer Drug Testing. Front. Oncol. 2022, 12, 976065. [Google Scholar] [CrossRef]
  121. Fiebig, H.H.; Schuchhardt, C.; Henss, H.; Fiedler, L.; Löhr, G.W. Comparison of Tumor Response in Nude Mice and in the Patients. Behring Inst. Mitteilungen 1984, 74, 343–352. [Google Scholar]
  122. Stewart, E.L.; Mascaux, C.; Pham, N.-A.; Sakashita, S.; Sykes, J.; Kim, L.; Yanagawa, N.; Allo, G.; Ishizawa, K.; Wang, D.; et al. Clinical Utility of Patient-Derived Xenografts to Determine Biomarkers of Prognosis and Map Resistance Pathways in EGFR-Mutant Lung Adenocarcinoma. J. Clin. Oncol. 2015, 33, 2472–2480. [Google Scholar] [CrossRef]
  123. Hidalgo, M.; Bruckheimer, E.; Rajeshkumar, N.; Garrido-Laguna, I.; De Oliveira, E.; Rubio-Viqueira, B.; Strawn, S.; Wick, M.J.; Martell, J.; Sidransky, D. A Pilot Clinical Study of Treatment Guided by Personalized Tumorgrafts in Patients with Advanced Cancer. Mol. Cancer Ther. 2011, 10, 1311–1316. [Google Scholar] [CrossRef] [PubMed]
  124. Stebbing, J.; Paz, K.; Schwartz, G.K.; Wexler, L.H.; Maki, R.; Pollock, R.E.; Morris, R.; Cohen, R.; Shankar, A.; Blackman, G.; et al. Patient-Derived Xenografts for Individualized Care in Advanced Sarcoma. Cancer 2014, 120, 2006–2015. [Google Scholar] [CrossRef] [PubMed]
  125. Sorokin, A.V.; Marie, P.K.; Bitner, L.; Syed, M.; Woods, M.; Manyam, G.; Kwong, L.N.; Johnson, B.; Morris, V.K.; Jones, P.; et al. Targeting Ras Mutant Colorectal Cancer with Dual Inhibition of Mek and Cdk4/6. Cancer Res. 2022, 82, 3335–3344. [Google Scholar] [CrossRef]
  126. Juric, D.; Castel, P.; Griffith, M.; Griffith, O.L.; Won, H.H.; Ellis, H.; Ebbesen, S.H.; Ainscough, B.J.; Ramu, A.; Iyer, G.; et al. Convergent Loss of Pten Leads to Clinical Resistance to a Pi(3)Kalpha Inhibitor. Nature 2015, 518, 240–244. [Google Scholar] [CrossRef] [PubMed]
  127. Rehman, S.K.; Haynes, J.; Collignon, E.; Brown, K.R.; Wang, Y.; Nixon, A.M.; Bruce, J.P.; Wintersinger, J.A.; Mer, A.S.; Lo, E.B.; et al. Colorectal Cancer Cells Enter a Diapause-like DTP State to Survive Chemotherapy. Cell 2021, 184, 226–242.e21. [Google Scholar] [CrossRef]
  128. Ocana, A.; Pandiella, A.; Siu, L.L.; Tannock, I.F. Preclinical Development of Molecular-Targeted Agents for Cancer. Nat. Rev. Clin. Oncol. 2010, 8, 200–209. [Google Scholar] [CrossRef]
  129. Patton, E.E.; Mueller, K.L.; Adams, D.J.; Anandasabapathy, N.; Aplin, A.E.; Bertolotto, C.; Bosenberg, M.; Ceol, C.J.; Burd, C.E.; Chi, P.; et al. Melanoma Models for the Next Generation of Therapies. Cancer Cell 2021, 39, 610–631. [Google Scholar] [CrossRef]
  130. Chuprin, J.; Buettner, H.; Seedhom, M.O.; Greiner, D.L.; Keck, J.G.; Ishikawa, F.; Shultz, L.D.; Brehm, M.A. Humanized Mouse Models for Immuno-Oncology Research. Nat. Rev. Clin. Oncol. 2023, 20, 192–206. [Google Scholar] [CrossRef]
  131. Choi, Y.; Lee, S.; Kim, K.; Kim, S.-H.; Chung, Y.-J.; Lee, C. Studying Cancer Immunotherapy Using Patient-Derived Xenografts (Pdxs) in Humanized Mice. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
  132. Marin-Jimenez, J.A.; Capasso, A.; Lewis, M.S.; Bagby, S.M.; Hartman, S.J.; Shulman, J.; Navarro, N.M.; Yu, H.; Rivard, C.J.; Wang, X.; et al. Testing Cancer Immunotherapy in a Human Immune System Mouse Model: Correlating Treatment Responses to Human Chimerism, Therapeutic Variables and Immune Cell Phenotypes. Front. Immunol. 2021, 12, 607282. [Google Scholar] [CrossRef] [PubMed]
  133. Martinov, T.; McKenna, K.M.; Tan, W.H.; Collins, E.J.; Kehret, A.R.; Linton, J.D.; Olsen, T.M.; Shobaki, N.; Rongvaux, A. Building the Next Generation of Humanized Hemato-Lymphoid System Mice. Front. Immunol. 2021, 12, 643852. [Google Scholar] [CrossRef] [PubMed]
  134. Legrand, N.; Huntington, N.D.; Nagasawa, M.; Bakker, A.Q.; Schotte, R.; Strick-Marchand, H.; de Geus, S.J.; Pouw, S.M.; Bohne, M.; Voordouw, A.; et al. Functional Cd47/Signal Regulatory Protein Alpha (Sirp(Alpha)) Interaction Is Required for Optimal Human T- and Natural Killer- (Nk) Cell Homeostasis in Vivo. Proc. Natl. Acad. Sci. USA 2011, 108, 13224–13229. [Google Scholar] [CrossRef] [PubMed]
  135. Jinnouchi, F.; Yamauchi, T.; Yurino, A.; Nunomura, T.; Nakano, M.; Iwamoto, C.; Obara, T.; Miyawaki, K.; Kikushige, Y.; Kato, K.; et al. A Human Sirpa Knock-in Xenograft Mouse Model to Study Human Hematopoietic and Cancer Stem Cells. Blood 2020, 135, 1661–1672. [Google Scholar] [CrossRef]
  136. Lavender, K.J.; Pang, W.W.; Messer, R.J.; Duley, A.K.; Race, B.; Phillips, K.; Scott, D.; Peterson, K.E.; Chan, C.K.; Dittmer, U.; et al. Blt-Humanized C57bl/6 Rag2-/-Gammac-/-Cd47-/- Mice Are Resistant to Gvhd and Develop B- and T-Cell Immunity to Hiv Infection. Blood 2013, 122, 4013–4020. [Google Scholar] [CrossRef]
  137. Hayakawa, J.; Hsieh, M.M.; Uchida, N.; Phang, O.; Tisdale, J.F. Busulfan Produces Efficient Human Cell Engraftment in Nod/Ltsz-Scid Il2rgamma(Null) Mice. Stem Cells 2009, 27, 175–182. [Google Scholar] [CrossRef]
  138. McIntosh, B.E.; Brown, M.E.; Duffin, B.M.; Maufort, J.P.; Vereide, D.T.; Slukvin; Thomson, J.A. Nonirradiated Nod,B6.Scid Il2rgamma-/- Kit(W41/W41) (Nbsgw) Mice Support Multilineage Engraftment of Human Hematopoietic Cells. Stem Cell Rep. 2015, 4, 171–180. [Google Scholar] [CrossRef]
  139. Rahmig, S.; Kronstein-Wiedemann, R.; Fohgrub, J.; Kronstein, N.; Nevmerzhitskaya, A.; Bornhäuser, M.; Gassmann, M.; Platz, A.; Ordemann, R.; Tonn, T.; et al. Improved Human Erythropoiesis and Platelet Formation in Humanized NSGW41 Mice. Stem Cell Rep. 2016, 7, 591–601. [Google Scholar] [CrossRef]
  140. Yurino, A.; Takenaka, K.; Yamauchi, T.; Nunomura, T.; Uehara, Y.; Jinnouchi, F.; Miyawaki, K.; Kikushige, Y.; Kato, K.; Miyamoto, T.; et al. Enhanced Reconstitution of Human Erythropoiesis and Thrombopoiesis in an Immunodeficient Mouse Model with Kit Wv Mutations. Stem Cell Rep. 2016, 7, 425–438. [Google Scholar] [CrossRef]
  141. Shan, L.; Flavell, R.A.; Herndler-Brandstetter, D. Development of Humanized Mouse Models for Studying Human NK Cells in Health and Disease. Methods Mol. Biol. 2022, 2463, 53–66. [Google Scholar]
  142. Rongvaux, A.; Takizawa, H.; Strowig, T.; Willinger, T.; Eynon, E.E.; Flavell, R.A.; Manz, M.G. Human Hemato-Lymphoid System Mice: Current Use and Future Potential for Medicine. Annu. Rev. Immunol. 2013, 31, 635–674. [Google Scholar] [CrossRef] [PubMed]
  143. Holyoake, T.L.; Nicolini, F.E.; Eaves, C.J. Functional Differences between Transplantable Human Hematopoietic Stem Cells from Fetal Liver, Cord Blood, and Adult Marrow. Exp. Hematol. 1999, 27, 1418–1427. [Google Scholar] [CrossRef] [PubMed]
  144. Brehm, M.A.; Kenney, L.L.; Wiles, M.V.; Low, B.E.; Tisch, R.M.; Burzenski, L.; Mueller, C.; Greiner, D.L.; Shultz, L.D. Lack of Acute Xenogeneic Graft- Versus-Host Disease, but Retention of T-Cell Function Following Engraftment of Human Peripheral Blood Mononuclear Cells in Nsg Mice Deficient in Mhc Class I and Ii Expression. FASEB J. 2019, 33, 3137–3151. [Google Scholar] [CrossRef] [PubMed]
  145. Yaguchi, T.; Kobayashi, A.; Inozume, T.; Morii, K.; Nagumo, H.; Nishio, H.; Iwata, T.; Ka, Y.; Katano, I.; Ito, R.; et al. Human Pbmc-Transferred Murine Mhc Class I/Ii-Deficient Nog Mice Enable Long-Term Evaluation of Human Immune Responses. Cell Mol. Immunol. 2018, 15, 953–962. [Google Scholar] [CrossRef] [PubMed]
  146. Zeng, Y.; Liu, B.; Rubio, M.T.; Wang, X.; Ojcius, D.M.; Tang, R.; Durrbach, A.; Ru, Z.; Zhou, Y.; Lone, Y.C. Creation of an Immunodeficient Hla-Transgenic Mouse (Humamice) and Functional Validation of Human Immunity after Transfer of Hla-Matched Human Cells. PLoS ONE 2017, 12, e0173754. [Google Scholar] [CrossRef]
  147. Holguin, L.; Echavarria, L.; Burnett, J.C. Novel Humanized Peripheral Blood Mononuclear Cell Mouse Model with Delayed Onset of Graft-Versus-Host Disease for Preclinical Hiv Research. J. Virol. 2022, 96, e0139421. [Google Scholar] [CrossRef]
  148. Strowig, T.; Gurer, C.; Ploss, A.; Liu, Y.F.; Arrey, F.; Sashihara, J.; Koo, G.; Rice, C.M.; Young, J.W.; Chadburn, A.; et al. Priming of Protective T Cell Responses against Virus-Induced Tumors in Mice with Human Immune System Components. J. Exp. Med. 2009, 206, 1423–1434. [Google Scholar] [CrossRef]
  149. Sippel, T.R.; Radtke, S.; Olsen, T.M.; Kiem, H.-P.; Rongvaux, A. Human Hematopoietic Stem Cell Maintenance and Myeloid Cell Development in Next-Generation Humanized Mouse Models. Blood Adv. 2019, 3, 268–274. [Google Scholar] [CrossRef]
  150. Ito, R.; Takahashi, T.; Katano, I.; Kawai, K.; Kamisako, T.; Ogura, T.; Ida-Tanaka, M.; Suemizu, H.; Nunomura, S.; Ra, C.; et al. Establishment of a Human Allergy Model Using Human Il-3/Gm-Csf-Transgenic Nog Mice. J. Immunol. 2013, 191, 2890–2899. [Google Scholar] [CrossRef]
  151. Yu, H.; Borsotti, C.; Schickel, J.N.; Zhu, S.; Strowig, T.; Eynon, E.E.; Frleta, D.; Gurer, C.; Murphy, A.J.; Yancopoulos, G.D.; et al. A Novel Humanized Mouse Model with Significant Improvement of Class-Switched, Antigen-Specific Antibody Production. Blood 2017, 129, 959–969. [Google Scholar] [CrossRef] [PubMed]
  152. Ono, R.; Watanabe, T.; Kawakami, E.; Iwasaki, M.; Tomizawa-Murasawa, M.; Matsuda, M.; Najima, Y.; Takagi, S.; Fujiki, S.; Sato, R.; et al. Co-Activation of Macrophages and T Cells Contribute to Chronic Gvhd in Human Il-6 Transgenic Humanised Mouse Model. Ebiomedicine 2019, 41, 584–596. [Google Scholar] [CrossRef] [PubMed]
  153. Lang, J.; Zhang, B.; Kelly, M.; Peterson, J.N.; Barbee, J.; Freed, B.M.; Di Santo, J.P.; Matsuda, J.L.; Torres, R.M.; Pelanda, R. Replacing Mouse Baff with Human Baff Does Not Improve B-Cell Maturation in Hematopoietic Humanized Mice. Blood Adv. 2017, 1, 2729–2741. [Google Scholar] [CrossRef] [PubMed]
  154. Lee, E.-C.; Liang, Q.; Ali, H.; Bayliss, L.; Beasley, A.; Bloomfield-Gerdes, T.; Bonoli, L.; Brown, R.; Campbell, J.; Carpenter, A.; et al. Complete Humanization of the Mouse Immunoglobulin Loci Enables Efficient Therapeutic Antibody Discovery. Nat. Biotechnol. 2014, 32, 356–363. [Google Scholar] [CrossRef]
  155. Kennedy, M.K.; Glaccum, M.; Brown, S.N.; Butz, E.A.; Viney, J.L.; Embers, M.; Matsuki, N.; Charrier, K.; Sedger, L.; Willis, C.R.; et al. Reversible Defects in Natural Killer and Memory Cd8 T Cell Lineages in Interleukin 15–Deficient Mice. J. Exp. Med. 2000, 191, 771–780. [Google Scholar] [CrossRef]
  156. Katano, I.; Takahashi, T.; Ito, R.; Kamisako, T.; Mizusawa, T.; Ka, Y.; Ogura, T.; Suemizu, H.; Kawakami, Y.; Ito, M. Predominant Development of Mature and Functional Human NK Cells in a Novel Human IL-2–Producing Transgenic NOG Mouse. J. Immunol. 2015, 194, 3513–3525. [Google Scholar] [CrossRef]
  157. Wunderlich, M.; Brooks, R.A.; Panchal, R.; Rhyasen, G.W.; Danet-Desnoyers, G.; Mulloy, J.C. Okt3 Prevents Xenogeneic Gvhd and Allows Reliable Xenograft Initiation from Unfractionated Human Hematopoietic Tissues. Blood 2014, 123, e134–e144. [Google Scholar] [CrossRef]
  158. Shultz, L.D.; Keck, J.; Burzenski, L.; Jangalwe, S.; Vaidya, S.; Greiner, D.L.; Brehm, M.A. Humanized Mouse Models of Immunological Diseases and Precision Medicine. Mamm. Genome 2019, 30, 123–142. [Google Scholar] [CrossRef]
  159. Wunderlich, M.; Chou, F.-S.; Sexton, C.; Presicce, P.; Chougnet, C.A.; Aliberti, J.; Mulloy, J.C. Improved Multilineage Human Hematopoietic Reconstitution and Function in Nsgs Mice. PLoS ONE 2018, 13, e0209034. [Google Scholar] [CrossRef]
  160. Lopez-Lastra, S.; Masse-Ranson, G.; Fiquet, O.; Darche, S.; Serafini, N.; Li, Y.; Dusséaux, M.; Strick-Marchand, H.; Di Santo, J.P. A Functional Dc Cross Talk Promotes Human Ilc Homeostasis in Humanized Mice. Blood Adv. 2017, 1, 601–614. [Google Scholar] [CrossRef]
  161. Li, Y.; Mention, J.-J.; Court, N.; Masse-Ranson, G.; Toubert, A.; Spits, H.; Legrand, N.; Corcuff, E.; Strick-Marchand, H.; Di Santo, J.P. A Novel Flt3-Deficient His Mouse Model with Selective Enhancement of Human Dc Development. Eur. J. Immunol. 2016, 46, 1291–1299. [Google Scholar] [CrossRef] [PubMed]
  162. Zheng, Y.; Sefik, E.; Astle, J.; Karatepe, K.; Öz, H.H.; Solis, A.G.; Jackson, R.; Luo, H.R.; Bruscia, E.M.; Halene, S.; et al. Human Neutrophil Development and Functionality Are Enabled in a Humanized Mouse Model. Proc. Natl. Acad. Sci. USA 2022, 119, e2121077119. [Google Scholar] [CrossRef] [PubMed]
  163. Ito, R.; Katano, I.; Kwok, I.W.; Ng, L.G.; Ida-Tanaka, M.; Ohno, Y.; Mu, Y.; Morita, H.; Nishinaka, E.; Nishime, C.; et al. Efficient Differentiation of Human Neutrophils with Recapitulation of Emergency Granulopoiesis in Human G-Csf Knockin Humanized Mice. Cell Rep. 2022, 41, 111841. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, Q.; Khoury, M.; Chen, J. Expression of Human Cytokines Dramatically Improves Reconstitution of Specific Human-Blood Lineage Cells in Humanized Mice. Proc. Natl. Acad. Sci. USA 2009, 106, 21783–21788. [Google Scholar] [CrossRef]
  165. Wang, Y.; Liu, S.; Yang, Z.; Algazi, A.P.; Lomeli, S.H.; Wang, Y.; Othus, M.; Hong, A.; Wang, X.; Randolph, C.E.; et al. Anti-Pd-1/L1 Lead-in before Mapk Inhibitor Combination Maximizes Antitumor Immunity and Efficacy. Cancer Cell 2021, 39, 1375–1387.e6. [Google Scholar] [CrossRef]
  166. Haas, L.; Elewaut, A.; Gerard, C.L.; Umkehrer, C.; Leiendecker, L.; Pedersen, M.; Krecioch, I.; Hoffmann, D.; Novatchkova, M.; Kuttke, M.; et al. Acquired Resistance to Anti-Mapk Targeted Therapy Confers an Immune-Evasive Tumor Microenvironment and Cross-Resistance to Immunotherapy in Melanoma. Nat. Cancer 2021, 2, 693–708. [Google Scholar] [CrossRef]
  167. Barrett, D.M.; Zhao, Y.; Liu, X.; Jiang, S.; Carpenito, C.; Carroll, R.G.; June, C.H.; Grupp, S.A.; Ramos, C.A.; Heslop, H.E.; et al. Treatment of Advanced Leukemia in Mice with mRNA Engineered T Cells. Hum. Gene Ther. 2011, 22, 1575–1586. [Google Scholar] [CrossRef]
  168. Schewe, D.M.; Alsadeq, A.; Sattler, C.; Lenk, L.; Vogiatzi, F.; Cario, G.; Vieth, S.; Valerius, T.; Rosskopf, S.; Meyersieck, F.; et al. An Fc-Engineered Cd19 Antibody Eradicates Mrd in Patient-Derived Mll-Rearranged Acute Lymphoblastic Leukemia Xenografts. Blood 2017, 130, 1543–1552. [Google Scholar] [CrossRef]
  169. Winterberg, D.; Lenk, L.; Oßwald, M.; Vogiatzi, F.; Gehlert, C.L.; Frielitz, F.-S.; Klausz, K.; Rösner, T.; Valerius, T.; Trauzold, A.; et al. Engineering of CD19 Antibodies: A CD19-TRAIL Fusion Construct Specifically Induces Apoptosis in B-Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) Cells In Vivo. J. Clin. Med. 2021, 10, 2634. [Google Scholar] [CrossRef]
  170. Wang, M.; Yao, L.C.; Cheng, M.; Cai, D.; Martinek, J.; Pan, C.X.; Shi, W.; Ma, A.H.; De Vere White, R.W.; Airhart, S.; et al. Humanized Mice in Studying Efficacy and Mechanisms of Pd-1-Targeted Cancer Immunotherapy. FASEB J. 2018, 32, 1537–1549. [Google Scholar] [CrossRef]
  171. Matas-Cespedes, A.; Vidal-Crespo, A.; Rodriguez, V.; Villamor, N.; Delgado, J.; Gine, E.; Roca-Ho, H.; Menendez, P.; Campo, E.; Lopez-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] [PubMed]
  172. Choi, B.; Lee, J.S.; Kim, S.J.; Hong, D.; Park, J.B.; Lee, K.-Y. Anti-Tumor Effects of Anti-Pd-1 Antibody, Pembrolizumab, in Humanized Nsg Pdx Mice Xenografted with Dedifferentiated Liposarcoma. Cancer Lett. 2020, 478, 56–69. [Google Scholar] [CrossRef] [PubMed]
  173. Cao, B.; Liu, M.; Huang, J.; Zhou, J.; Li, J.; Lian, H.; Huang, W.; Guo, Y.; Yang, S.; Lin, L.; et al. Development of Mesothelin-Specific Car Nk-92 Cells for the Treatment of Gastric Cancer. Int. J. Biol. Sci. 2021, 17, 3850–3861. [Google Scholar] [CrossRef]
  174. Zhao, Y.; Shuen, T.W.H.; Toh, T.B.; Chan, X.Y.; Liu, M.; Tan, S.Y.; Fan, Y.; Yang, H.; Lyer, S.G.; Bonney, G.K.; et al. Development of a New Patient-Derived Xenograft Humanised Mouse Model to Study Human-Specific Tumour Microenvironment and Immunotherapy. Gut 2018, 67, 1845–1854. [Google Scholar] [CrossRef] [PubMed]
  175. Kundu, K.; Ghosh, S.; Sarkar, R.; Edri, A.; Brusilovsky, M.; Gershoni-Yahalom, O.; Yossef, R.; Shemesh, A.; Soria, J.C.; Lazar, V.; et al. Inhibition of the Nkp44-Pcna Immune Checkpoint Using a Mab to Pcna. Cancer Immunol. Res. 2019, 7, 1120–1134. [Google Scholar] [CrossRef] [PubMed]
  176. Barry, W.E.; Jackson, J.R.; Asuelime, G.E.; Wu, H.W.; Sun, J.; Wan, Z.; Malvar, J.; Sheard, M.A.; Wang, L.; Seeger, R.C.; et al. Activated Natural Killer Cells in Combination with Anti-Gd2 Antibody Dinutuximab Improve Survival of Mice after Surgical Resection of Primary Neuroblastoma. Clin. Cancer Res. 2019, 25, 325–333. [Google Scholar] [CrossRef]
  177. Gitto, S.B.; Kim, H.; Rafail, S.; Omran, D.K.; Medvedev, S.; Kinose, Y.; Rodriguez-Garcia, A.; Flowers, A.J.; Xu, H.; Schwartz, L.E.; et al. An Autologous Humanized Patient-Derived-Xenograft Platform to Evaluate Immunotherapy in Ovarian Cancer. Gynecol. Oncol. 2020, 156, 222–232. [Google Scholar] [CrossRef]
  178. Sommaggio, R.; Cappuzzello, E.; Pietà, A.D.; Tosi, A.; Palmerini, P.; Carpanese, D.; Nicolè, L.; Rosato, A. Adoptive Cell Therapy of Triple Negative Breast Cancer with Redirected Cytokine-Induced Killer Cells. Oncoimmunology 2020, 9, 1777046. [Google Scholar] [CrossRef]
  179. Ku, A.; Kondo, M.; Cai, Z.; Meens, J.; Li, M.R.; Ailles, L.; Reilly, R.M. Dose Predictions for [(177)Lu]Lu-Dota-Panitumumab F(Ab’)(2) in Nrg Mice with Hnscc Patient-Derived Tumour Xenografts Based on [(64)Cu]Cu-Dota-Panitumumab F(Ab’)(2)—Implications for a Pet Theranostic Strategy. EJNMMI Radiopharm. Chem. 2021, 6, 25. [Google Scholar] [CrossRef]
  180. Park, J.A.; Cheung, N.-K.V. Gd2 or Her2 Targeting T Cell Engaging Bispecific Antibodies to Treat Osteosarcoma. J. Hematol. Oncol. 2020, 13, 172. [Google Scholar] [CrossRef]
  181. Lang, J.; Leal, A.D.; Marín-Jiménez, J.A.; Hartman, S.J.; Shulman, J.; Navarro, N.M.; Lewis, M.S.; Capasso, A.; Bagby, S.M.; Yacob, B.W.; et al. Cabozantinib Sensitizes Microsatellite Stable Colorectal Cancer to Immune Checkpoint Blockade by Immune Modulation in Human Immune System Mouse Models. Front. Oncol. 2022, 12, 877635. [Google Scholar] [CrossRef]
  182. Capasso, A.; Lang, J.; Pitts, T.M.; Jordan, K.R.; Lieu, C.H.; Davis, S.L.; Diamond, J.R.; Kopetz, S.; Barbee, J.; Peterson, J.; et al. Characterization of Immune Responses to Anti-Pd-1 Mono and Combination Immunotherapy in Hematopoietic Humanized Mice Implanted with Tumor Xenografts. J. Immunother. Cancer 2019, 7, 37. [Google Scholar] [CrossRef] [PubMed]
  183. Lang, J.; Capasso, A.; Jordan, K.R.; French, J.D.; Kar, A.; Bagby, S.M.; Barbee, J.; Yacob, B.W.; Head, L.S.; Tompkins, K.D.; et al. Development of an Adrenocortical Cancer Humanized Mouse Model to Characterize Anti-PD1 Effects on Tumor Microenvironment. J. Clin. Endocrinol. Metab. 2020, 105, 26–42. [Google Scholar] [CrossRef]
  184. Jespersen, H.; Lindberg, M.F.; Donia, M.; Söderberg, E.M.V.; Andersen, R.; Keller, U.; Ny, L.; Svane, I.M.; Nilsson, L.M.; Nilsson, J.A. Clinical Responses to Adoptive T-Cell Transfer Can Be Modeled in an Autologous Immune-Humanized Mouse Model. Nat. Commun. 2017, 8, 707. [Google Scholar] [CrossRef]
  185. Forsberg, E.M.; Lindberg, M.F.; Jespersen, H.; Alsén, S.; Bagge, R.O.; Donia, M.; Svane, I.M.; Nilsson, O.; Ny, L.; Nilsson, L.M.; et al. HER2 CAR-T Cells Eradicate Uveal Melanoma and T-cell Therapy–Resistant Human Melanoma in IL2 Transgenic NOD/SCID IL2 Receptor Knockout Mice. Cancer Res 2019, 79, 899–904. [Google Scholar] [CrossRef]
  186. Maser, I.-P.; Hoves, S.; Bayer, C.; Heidkamp, G.; Nimmerjahn, F.; Eckmann, J.; Ries, C.H. The Tumor Milieu Promotes Functional Human Tumor-Resident Plasmacytoid Dendritic Cells in Humanized Mouse Models. Front. Immunol. 2020, 11, 2082. [Google Scholar] [CrossRef] [PubMed]
  187. Wunderlich, M.; Manning, N.; Sexton, C.; O’brien, E.; Byerly, L.; Stillwell, C.; Perentesis, J.P.; Mulloy, J.C.; Mizukawa, B. PD-1 Inhibition Enhances Blinatumomab Response in a UCB/PDX Model of Relapsed Pediatric B-Cell Acute Lymphoblastic Leukemia. Front. Oncol. 2021, 11, 642466. [Google Scholar] [CrossRef]
  188. Myburgh, R.; Kiefer, J.D.; Russkamp, N.F.; Magnani, C.F.; Nuñez, N.; Simonis, A.; Pfister, S.; Wilk, C.M.; McHugh, D.; Friemel, J.; et al. Anti-Human Cd117 Car T-Cells Efficiently Eliminate Healthy and Malignant Cd117-Expressing Hematopoietic Cells. Leukemia 2020, 34, 2688–2703. [Google Scholar] [CrossRef] [PubMed]
  189. Biernacki, M.A.; Foster, K.A.; Woodward, K.B.; Coon, M.E.; Cummings, C.; Cunningham, T.M.; Dossa, R.G.; Brault, M.; Stokke, J.; Olsen, T.M.; et al. Cbfb-Myh11 Fusion Neoantigen Enables T Cell Recognition and Killing of Acute Myeloid Leukemia. J. Clin. Investig. 2020, 130, 5127–5141. [Google Scholar] [CrossRef]
  190. Nguyen, R.; Patel, A.; Griffiths, L.M.; Dapper, J.; Stewart, E.A.; Houston, J.; Johnson, M.; Akers, W.J.; Furman, W.L.; Dyer, M.A. Next-Generation Humanized Patient-Derived Xenograft Mouse Model for Pre-Clinical Antibody Studies in Neuroblastoma. Cancer Immunol. Immunother. 2021, 70, 721–732. [Google Scholar] [CrossRef]
  191. Hashwah, H.; Bertram, K.; Stirm, K.; Stelling, A.; Wu, C.T.; Kasser, S.; Manz, M.G.; Theocharides, A.P.; Tzankov, A.; Muller, A. The Il-6 Signaling Complex Is a Critical Driver, Negative Prognostic Factor, and Therapeutic Target in Diffuse Large B-Cell Lymphoma. EMBO Mol. Med. 2019, 11, e10576. [Google Scholar] [CrossRef] [PubMed]
  192. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated Regulation of Myeloid Cells by Tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed]
  193. Pittet, M.J.; Michielin, O.; Migliorini, D. Clinical Relevance of Tumour-Associated Macrophages. Nat. Rev. Clin. Oncol. 2022, 19, 402–421. [Google Scholar] [CrossRef] [PubMed]
  194. Bruni, D.; Angell, H.K.; Galon, J. The Immune Contexture and Immunoscore in Cancer Prognosis and Therapeutic Efficacy. Nat. Rev. Cancer 2020, 20, 662–680. [Google Scholar] [CrossRef] [PubMed]
  195. Ruffell, B.; Coussens, L.M. Macrophages and Therapeutic Resistance in Cancer. Cancer Cell 2015, 27, 462–472. [Google Scholar] [CrossRef] [PubMed]
  196. Cassetta, L.; Pollard, J.W. Targeting Macrophages: Therapeutic Approaches in Cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef]
  197. Mantovani, A.; Allavena, P.; Marchesi, F.; Garlanda, C. Macrophages as Tools and Targets in Cancer Therapy. Nat. Rev. Drug Discov. 2022, 21, 799–820. [Google Scholar] [CrossRef]
  198. Yu, C.I.; Martinek, J.; Wu, T.-C.; Kim, K.I.; George, J.; Ahmadzadeh, E.; Maser, R.; Marches, F.; Metang, P.; Authie, P.; et al. Human Kit+ Myeloid Cells Facilitate Visceral Metastasis by Melanoma. J. Exp. Med. 2021, 218, e20182163. [Google Scholar] [CrossRef]
  199. Su, S.; Liu, Q.; Chen, J.; Chen, J.; Chen, F.; He, C.; Huang, D.; Wu, W.; Lin, L.; Huang, W.; et al. A Positive Feedback Loop between Mesenchymal-like Cancer Cells and Macrophages Is Essential to Breast Cancer Metastasis. Cancer Cell 2014, 25, 605–620. [Google Scholar] [CrossRef]
  200. Rosato, R.R.; Dávila-González, D.; Choi, D.S.; Qian, W.; Chen, W.; Kozielski, A.J.; Wong, H.; Dave, B.; Chang, J.C. Evaluation of Anti-Pd-1-Based Therapy against Triple-Negative Breast Cancer Patient-Derived Xenograft Tumors Engrafted in Humanized Mouse Models. Breast Cancer Res. 2018, 20, 108. [Google Scholar] [CrossRef]
  201. Yao, L.-C.; Aryee, K.-E.; Cheng, M.; Kaur, P.; Keck, J.G.; Brehm, M.A. Creation of PDX-Bearing Humanized Mice to Study Immuno-oncology. Methods Mol. Biol. 2019, 1953, 241–252. [Google Scholar] [PubMed]
  202. Hodis, E.; Triglia, E.T.; Kwon, J.Y.H.; Biancalani, T.; Zakka, L.R.; Parkar, S.; Hutter, J.C.; Buffoni, L.; Delorey, T.M.; Phillips, D.; et al. Stepwise-Edited, Human Melanoma Models Reveal Mutations’ Effect on Tumor and Microenvironment. Science 2022, 376, eabi8175. [Google Scholar] [CrossRef] [PubMed]
  203. De Angelis, M.L.; Francescangeli, F.; Nicolazzo, C.; Xhelili, E.; La Torre, F.; Colace, L.; Bruselles, A.; Macchia, D.; Vitale, S.; Gazzaniga, P.; et al. An Orthotopic Patient-Derived Xenograft (PDX) Model Allows the Analysis of Metastasis-Associated Features in Colorectal Cancer. Front. Oncol. 2022, 12, 869485. [Google Scholar] [CrossRef] [PubMed]
  204. Jaiswal, S.; Jamieson, C.H.; Pang, W.W.; Park, C.Y.; Chao, M.P.; Majeti, R.; Traver, D.; van Rooijen, N.; Weissman, I.L. CD47 Is Upregulated on Circulating Hematopoietic Stem Cells and Leukemia Cells to Avoid Phagocytosis. Cell 2009, 138, 271–285. [Google Scholar] [CrossRef]
  205. Ring, N.G.; Herndler-Brandstetter, D.; Weiskopf, K.; Shan, L.; Volkmer, J.P.; George, B.M.; Lietzenmayer, M.; McKenna, K.M.; Naik, T.J.; McCarty, A.; et al. Anti-Sirpalpha Antibody Immunotherapy Enhances Neutrophil and Macrophage Antitumor Activity. Proc. Natl. Acad. Sci. USA 2017, 114, E10578–E10585. [Google Scholar] [CrossRef]
  206. Morton, J.J.; Alzofon, N.; Keysar, S.B.; Chimed, T.S.; Reisinger, J.; Perrenoud, L.; Le, P.N.; Nieto, C.; Gomez, K.; Miller, B.; et al. Studying Immunotherapy Resistance in a Melanoma Autologous Humanized Mouse Xenograft. Mol. Cancer Res. 2021, 19, 346–357. [Google Scholar] [CrossRef] [PubMed]
  207. Pyo, K.H.; Kim, J.H.; Lee, J.-M.; Kim, S.E.; Cho, J.S.; Lim, S.M.; Cho, B.C. Promising Preclinical Platform for Evaluation of Immuno-Oncology Drugs Using Hu-Pbl-Nsg Lung Cancer Models. Lung Cancer 2019, 127, 112–121. [Google Scholar] [CrossRef]
  208. Meraz, I.M.; Majidi, M.; Meng, F.; Shao, R.; Ha, M.J.; Neri, S.; Fang, B.; Lin, S.H.; Tinkey, P.T.; Shpall, E.J.; et al. An Improved Patient-Derived Xenograft Humanized Mouse Model for Evaluation of Lung Cancer Immune Responses. Cancer Immunol. Res. 2019, 7, 1267–1279. [Google Scholar] [CrossRef]
  209. Kang, Y.; Armstrong, A.J.; Hsu, D.S. An Autologous Humanized Patient-Derived Xenograft (Pdx) Model for Evaluation of Nivolumab Immunotherapy in Renal Cell Cancer: A Case Report. Stem Cell Investig. 2022, 9, 8. [Google Scholar] [CrossRef]
  210. Tentler, J.J.; Lang, J.; Capasso, A.; Kim, D.J.; Benaim, E.; Lee, Y.B.; Eisen, A.; Bagby, S.M.; Hartman, S.J.; Yacob, B.W.; et al. Rx-5902, a Novel Beta-Catenin Modulator, Potentiates the Efficacy of Immune Checkpoint Inhibitors in Preclinical Models of Triple-Negative Breast Cancer. BMC Cancer 2020, 20, 1063. [Google Scholar] [CrossRef]
  211. Somasundaram, R.; Connelly, T.; Choi, R.; Choi, H.; Samarkina, A.; Li, L.; Gregorio, E.; Chen, Y.; Thakur, R.; Abdel-Mohsen, M.; et al. Tumor-Infiltrating Mast Cells Are Associated with Resistance to Anti-Pd-1 Therapy. Nat. Commun. 2021, 12, 346. [Google Scholar] [CrossRef] [PubMed]
  212. Küçükköse, E.; Heesters, B.A.; Villaudy, J.; Verheem, A.; Cercel, M.; van Hal, S.; Boj, S.F.; Rinkes, I.H.M.B.; Punt, C.J.A.; Roodhart, J.M.L.; et al. Modeling Resistance of Colorectal Peritoneal Metastases to Immune Checkpoint Blockade in Humanized Mice. J. Immunother. Cancer 2022, 10, e005345. [Google Scholar] [CrossRef] [PubMed]
  213. Fourcade, J.; Sun, Z.; Benallaoua, M.; Guillaume, P.; Luescher, I.F.; Sander, C.; Kirkwood, J.M.; Kuchroo, V.; Zarour, H.M. Upregulation of Tim-3 and Pd-1 Expression Is Associated with Tumor Antigen-Specific Cd8+ T Cell Dysfunction in Melanoma Patients. J. Exp. Med. 2010, 207, 2175–2186. [Google Scholar] [CrossRef] [PubMed]
  214. Sakuishi, K.; Apetoh, L.; Sullivan, J.M.; Blazar, B.R.; Kuchroo, V.K.; Anderson, A.C. Targeting Tim-3 and Pd-1 Pathways to Reverse T Cell Exhaustion and Restore Anti-Tumor Immunity. J. Exp. Med. 2010, 207, 2187–2194. [Google Scholar] [CrossRef]
  215. Chhabra, Y.; Weeraratna, A.T. Fibroblasts in Cancer: Unity in Heterogeneity. Cell 2023, 186, 1580–1609. [Google Scholar] [CrossRef]
  216. Delitto, D.; Pham, K.; Vlada, A.C.; Sarosi, G.A.; Thomas, R.M.; Behrns, K.E.; Liu, C.; Hughes, S.J.; Wallet, S.M.; Trevino, J.G. Patient-Derived Xenograft Models for Pancreatic Adenocarcinoma Demonstrate Retention of Tumor Morphology through Incorporation of Murine Stromal Elements. Am. J. Pathol. 2015, 185, 1297–1303. [Google Scholar] [CrossRef]
  217. Karnoub, A.E.; Dash, A.B.; Vo, A.P.; Sullivan, A.; Brooks, M.W.; Bell, G.W.; Richardson, A.L.; Polyak, K.; Tubo, R.; Weinberg, R.A. Mesenchymal Stem Cells within Tumour Stroma Promote Breast Cancer Metastasis. Nature 2007, 449, 557–563. [Google Scholar] [CrossRef]
  218. Cho, J.; Lee, H.-J.; Hwang, S.J.; Min, H.-Y.; Kang, H.N.; Park, A.-Y.; Hyun, S.Y.; Sim, J.Y.; Lee, H.J.; Jang, H.-J.; et al. The Interplay between Slow-Cycling, Chemoresistant Cancer Cells and Fibroblasts Creates a Proinflammatory Niche for Tumor Progression. Cancer Res. 2020, 80, 2257–2272. [Google Scholar] [CrossRef]
  219. Han, J.; Yun, J.; Quan, M.; Kang, W.; Jung, J.-G.; Heo, W.; Li, S.; Lee, K.J.; Son, H.-Y.; Kim, J.H.; et al. Jak2 Regulates Paclitaxel Resistance in Triple Negative Breast Cancers. J. Mol. Med. 2021, 99, 1783–1795. [Google Scholar] [CrossRef]
Figure 1. Humanized mouse models for translational cancer research and immunotherapy. The scheme illustrates the genealogical tree of different humanized mouse strains. Details on the characteristics of the individual strains and their usability for generating PDXs and for testing cancer immunotherapies can be found in the text and are summarized in Table 1 and Table 2. Humanized mouse strains that are not commonly used or that are commercial regenerations of established mouse models (e.g., NCG, B-NDG) have not been included in this figure. NRG-A2/DR4 mice are also called “DRAGA” mice. Abbreviations: B6RG, C57BL/6 Rag2-/- Il2rg-/-; BRG, BALB/c Rag2-/- Il2rg-/-; NOD, non-obese diabetic; NOG, NOD SCID Il2rgnull; NRG, NOD Rag1-/- Il2rg-/-; NSG, NOD SCID Il2rg-/-; KI, knock-in; SCID, severe combined immunodeficiency; SRG, SIRPA Rag2-/- Il2rg-/-; MISTRG, M-CSF IL3/GM-CSF SIRPA TPO Rag2-/- Il2rg-/-; Tg, transgene.
Figure 1. Humanized mouse models for translational cancer research and immunotherapy. The scheme illustrates the genealogical tree of different humanized mouse strains. Details on the characteristics of the individual strains and their usability for generating PDXs and for testing cancer immunotherapies can be found in the text and are summarized in Table 1 and Table 2. Humanized mouse strains that are not commonly used or that are commercial regenerations of established mouse models (e.g., NCG, B-NDG) have not been included in this figure. NRG-A2/DR4 mice are also called “DRAGA” mice. Abbreviations: B6RG, C57BL/6 Rag2-/- Il2rg-/-; BRG, BALB/c Rag2-/- Il2rg-/-; NOD, non-obese diabetic; NOG, NOD SCID Il2rgnull; NRG, NOD Rag1-/- Il2rg-/-; NSG, NOD SCID Il2rg-/-; KI, knock-in; SCID, severe combined immunodeficiency; SRG, SIRPA Rag2-/- Il2rg-/-; MISTRG, M-CSF IL3/GM-CSF SIRPA TPO Rag2-/- Il2rg-/-; Tg, transgene.
Cancers 15 02989 g001
Figure 2. Modeling patient-derived tumors in immunodeficient mice. (A) Patient-derived tumor xenograft (PDX) models are established by implanting patient tumor into immunodeficient mice. The PDXs are expanded in vivo by serial passages to establish a PDX biobank. (B) Factors that impact the successful establishment of PDXs (initial growth and in vivo propagation for at least 5 passages). (C) Immunodeficient mouse models have unique characteristics (e.g., expression of human cytokines) and therefore differently support the engraftment of various cancer types. Abbreviations: AML, acute myeloid leukemia; APL, acute promyelocytic leukemia; CAR, chimeric antigen receptor; CNA, copy number alteration; HR+, hormone receptor-positive; IL-6, interleukin 6; M-CSF, macrophage colony-stimulating factor; MDS, myelodysplastic syndrome; MM, multiple myeloma.
Figure 2. Modeling patient-derived tumors in immunodeficient mice. (A) Patient-derived tumor xenograft (PDX) models are established by implanting patient tumor into immunodeficient mice. The PDXs are expanded in vivo by serial passages to establish a PDX biobank. (B) Factors that impact the successful establishment of PDXs (initial growth and in vivo propagation for at least 5 passages). (C) Immunodeficient mouse models have unique characteristics (e.g., expression of human cytokines) and therefore differently support the engraftment of various cancer types. Abbreviations: AML, acute myeloid leukemia; APL, acute promyelocytic leukemia; CAR, chimeric antigen receptor; CNA, copy number alteration; HR+, hormone receptor-positive; IL-6, interleukin 6; M-CSF, macrophage colony-stimulating factor; MDS, myelodysplastic syndrome; MM, multiple myeloma.
Cancers 15 02989 g002
Figure 3. Establishment of human immune system (HIS) mouse models. (A) Immunodeficient mice can be reconstituted with a human immune system. For this purpose, newborn mice (1–5 days old) are preconditioned (e.g., sublethal irradiation, busulfan treatment). Some humanized mouse strains, such as NSG-W41, NRG-W41, B6RGSNOD-K, MISTRG and MISTRG-6 mice, do not need to be preconditioned. Human HSPCs are injected into the liver of newborn mice, which leads to the development of a human immune system. (B) The bar graph illustrates the composition of the human immune system in five different humanized mouse models 10–14 weeks post-engraftment with fetal/neonatal HSPCs [14,149,150]. The development, composition and function of the human immune system depends on the humanized mouse model and the source of HSPCs. Fetal and neonatal HSPCs (fetal liver, umbilical cord blood) engraft ≥ 3-fold better than adult HSPCs (bone marrow, G-CSF-mobilized blood) [18,143]. (C) MHC-deficient humanized mice can be preconditioned and intravenously engrafted with human PBMCs. (D) The bar graph illustrates the composition of human immune system in NSG and MHC-deficient NSG mice 4 weeks post-engraftment with human PBMCs [144]. Abbreviations: cGy, centigray; G-CSF, granulocyte colony-stimulating factor; HSPC, hematopoietic stem and progenitor cell; Hu-PBL, human peripheral blood lymphocyte; MHC, major histocompatibility complex; NK cell, natural killer cell; PBMC, peripheral blood mononuclear cells.
Figure 3. Establishment of human immune system (HIS) mouse models. (A) Immunodeficient mice can be reconstituted with a human immune system. For this purpose, newborn mice (1–5 days old) are preconditioned (e.g., sublethal irradiation, busulfan treatment). Some humanized mouse strains, such as NSG-W41, NRG-W41, B6RGSNOD-K, MISTRG and MISTRG-6 mice, do not need to be preconditioned. Human HSPCs are injected into the liver of newborn mice, which leads to the development of a human immune system. (B) The bar graph illustrates the composition of the human immune system in five different humanized mouse models 10–14 weeks post-engraftment with fetal/neonatal HSPCs [14,149,150]. The development, composition and function of the human immune system depends on the humanized mouse model and the source of HSPCs. Fetal and neonatal HSPCs (fetal liver, umbilical cord blood) engraft ≥ 3-fold better than adult HSPCs (bone marrow, G-CSF-mobilized blood) [18,143]. (C) MHC-deficient humanized mice can be preconditioned and intravenously engrafted with human PBMCs. (D) The bar graph illustrates the composition of human immune system in NSG and MHC-deficient NSG mice 4 weeks post-engraftment with human PBMCs [144]. Abbreviations: cGy, centigray; G-CSF, granulocyte colony-stimulating factor; HSPC, hematopoietic stem and progenitor cell; Hu-PBL, human peripheral blood lymphocyte; MHC, major histocompatibility complex; NK cell, natural killer cell; PBMC, peripheral blood mononuclear cells.
Cancers 15 02989 g003
Figure 4. Cancer immunotherapy in humanized PDX mouse models. Efficient and functional development of human immune cell lineages depends on the humanized mouse model. This also influences the composition of the TIME and the suitability of investigating different types of immunotherapies [14,18,83]. Abbreviations: ADCC, antibody-dependent cellular cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; CTLA-4, cytotoxic T lymphocyte-associated antigen 4; HIS, human immune system; KIT (also known as c-kit, CD117 or stem cell factor receptor); NK, natural killer; PD-1, programmed cell death protein 1; TAM, tumor-associated macrophage; TIME, tumor immune microenvironment; VEGF, vascular endothelial growth factor.
Figure 4. Cancer immunotherapy in humanized PDX mouse models. Efficient and functional development of human immune cell lineages depends on the humanized mouse model. This also influences the composition of the TIME and the suitability of investigating different types of immunotherapies [14,18,83]. Abbreviations: ADCC, antibody-dependent cellular cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; CTLA-4, cytotoxic T lymphocyte-associated antigen 4; HIS, human immune system; KIT (also known as c-kit, CD117 or stem cell factor receptor); NK, natural killer; PD-1, programmed cell death protein 1; TAM, tumor-associated macrophage; TIME, tumor immune microenvironment; VEGF, vascular endothelial growth factor.
Cancers 15 02989 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chen, A.; Neuwirth, I.; Herndler-Brandstetter, D. Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice. Cancers 2023, 15, 2989. https://doi.org/10.3390/cancers15112989

AMA Style

Chen A, Neuwirth I, Herndler-Brandstetter D. Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice. Cancers. 2023; 15(11):2989. https://doi.org/10.3390/cancers15112989

Chicago/Turabian Style

Chen, Anna, Ines Neuwirth, and Dietmar Herndler-Brandstetter. 2023. "Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice" Cancers 15, no. 11: 2989. https://doi.org/10.3390/cancers15112989

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