Next Article in Journal
Diagnosing Ovarian Cancer on MRI: A Preliminary Study Comparing Deep Learning and Radiologist Assessments
Next Article in Special Issue
Molecular Mechanisms of Kaposi Sarcoma Development
Previous Article in Journal
[68Ga]DOTATOC PET/CT Radiomics to Predict the Response in GEP-NETs Undergoing [177Lu]DOTATOC PRRT: The “Theragnomics” Concept
Previous Article in Special Issue
Therapeutic Perspectives in the Systemic Treatment of Kaposi’s Sarcoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Reconstitution Inflammatory Syndrome Associated Kaposi Sarcoma

by
Isabelle Poizot-Martin
1,*,
Sylvie Brégigeon
2,
Romain Palich
3,
Anne-Geneviève Marcelin
4,
Marc-Antoine Valantin
3,
Caroline Solas
5,
Marianne Veyri
6,
Jean-Philippe Spano
6 and
Alain Makinson
7
1
Assistance Publique-Hôpitaux de Marseille (APHM), Inserm, Institut de Recherche pour le Développement (IRD), SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l’Information Médicale, ISSPAM, APHM Sainte-Marguerite, Service D’immuno-Hématologie Clinique, Aix-Marseille Université, 13009 Marseille, France
2
Assistance Publique-Hôpitaux de Marseille (APHM) Sainte-Marguerite, Service D’immuno-Hématologie Clinique, Aix-Marseille Université, 13009 Marseille, France
3
Department of Infectious Diseases, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Pierre Louis Epidemiology and Public Health Institute (iPLESP), INSERM U1136, Sorbonne University, 75013 Paris, France
4
INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique (iPLESP), AP-HP, Hôpital Pitié Salpêtrière, Service de Virologie, Sorbonne Université, 75013 Paris, France
5
Assistance Publique-Hôpitaux de Marseille (APHM), Hôpital La Timone, Laboratoire de Pharmacocinétique et Toxicologie, INSERM 1207, IRD 190, Unité des Virus Emergents, Aix-Marseille Université, 13005 Marseille, France
6
Department of Medical Oncology, Pitié Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Institut Universitaire de Cancérologie (IUC), CLIP2 Galilée, Pierre Louis Epidemiology and Public Health Institute (iPLESP), INSERM U1136, Sorbonne Université, 75013 Paris, France
7
Centre Hospitalier Universitaire de Montpellier, Département des Maladies Infectieuses et Tropicales, INSERM U1175/IRD UMI 233, 34000 Montpellier, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(4), 986; https://doi.org/10.3390/cancers14040986
Submission received: 29 November 2021 / Revised: 28 January 2022 / Accepted: 9 February 2022 / Published: 16 February 2022
(This article belongs to the Special Issue Perspectives on Kaposi's Sarcoma)

Abstract

:

Simple Summary

Kaposi sarcoma (KS) incidence has declined substantially since the advent of effective ART but it remains a frequent cancer among people living with HIV, including those on ART with a sustained undetectable HIV viral load and in late presenters. ART is responsible for KS improvement and resolution, but new onset (unmasking KS-IRIS) or sudden progression of preexisting KS (paradoxical KS-IRIS) can occur, even in patients with a low degree of immunocompromise. Both paradoxical and unmasking KS-IRIS have been associated with significant morbidity and mortality. We carried out a literature review regarding the incidence, pathogenic mechanisms, risk factors, clinical presentation, and management strategies of KS-IRIS in the ART era.

Abstract

People living with HIV (PLWH) with advanced immunosuppression who initiate antiretroviral therapy (ART) are susceptible to the occurrence of an immune reconstitution inflammatory syndrome (IRIS). Although ART is responsible for AIDS- associated Kaposi sarcoma (KS) improvement and resolution, new onset (unmasking KS-IRIS) or sudden progression of preexisting KS (paradoxical KS-IRIS) can occur after a time delay of between a few days and 6 months after the initiation or resumption of ART, even in patients with a low degree of immunocompromise. KS-IRIS incidence varies from 2.4% to 39%, depending on study design, populations, and geographic regions. Risk factors for developing KS-IRIS include advanced KS tumor stage (T1), pre-treatment HIV viral load >5 log10 copies/mL, detectable pre-treatment plasma-KSHV, and initiation of ART alone without concurrent chemotherapy. Both paradoxical and unmasking KS-IRIS have been associated with significant morbidity and mortality, and thrombocytopenia (<100,000 platelets/mm3 at 12 weeks) has been associated with death. KS-IRIS is not to be considered as ART failure, and an ART regimen must be pursued. Systemic chemotherapy for KS in conjunction with ART is recommended and, in contrast with management of IRIS for other opportunistic infections, glucocorticoids are contra-indicated. Despite our preliminary results, the place of targeted therapies in the prevention or treatment of KS-IRIS needs further assessment.

1. Introduction

People living with HIV (PLWH) and with advanced immunosuppression who initiate antiretroviral therapy (ART) are susceptible to the occurrence of an immune reconstitution inflammatory syndrome (IRIS) [1]. This syndrome results from the restoration of a dysregulated immune response after ART initiation, optimization or resumption [2], causing an exaggerated inflammatory response against persistent infectious or non-infectious pathogen-specific antigens [1,3,4]. Due to the persistence of a high proportion of late presenters at the time of HIV diagnosis, including in countries with universal access to ART [5], IRIS risk still remains, increasing the burden of morbidity and mortality [1,2].
A wide spectrum of clinical manifestations can be observed during IRIS with two distinct temporal patterns recognized as (1) paradoxical IRIS, when there is a worsening or recurrence of previously treated opportunistic infections (OI) symptoms or other inflammatory auto-immune or neoplastic processes that occurs despite an earlier favorable response to therapy prior to ART, and (2) unmasking IRIS, when one or more of these events occurred following ART initiation [6,7,8,9].
KS has long remained the most common malignant condition in people living with HIV (PLWH) and is one of the most severe complications of full-blown AIDS.
KS is now recognized as a cytokine-mediated angioproliferative disease caused by a human herpesvirus [8] (HHV8), also referred as the Kaposi sarcoma-associated herpesvirus (KSHV) [10,11,12]. KSHV is an oncogenic virus belonging to the Herpesviridae family found in the lesions of all epidemiological forms of Kaposi’s sarcoma [13]. KSHV encodes oncogenic proteins that can modulate cellular pathways, leading to inhibition of apoptosis, cell proliferation stimulation, angiogenesis, inflammation, and immune escape, and therefore is involved in KS development [14,15].The widespread use of ART has modified the natural history and incidence of KS as well as its clinical features and prognosis in resource-rich countries more than in low-income countries, particularly in sub-Saharan Africa (SSA) where KS is still considered as a public health challenge [16,17].
In resource-rich countries, KS incidence has declined substantially since the advent of effective ART [18,19], but it remains a frequent cancer among PLWH [20], including those on ART with a sustained undetectable HIV viral load [21,22,23] and in late presenters, as observed between 2010 and 2015 in the French Dat’AIDS cohort [24]. Furthermore, KS is still an important cause of death [25]. Although ART is responsible for KS improvement and resolution, the new onset or sudden progression of preexisting Kaposi sarcoma (KS) within 3 months of the initiation of ART is a well described event among PLWH [26,27,28,29].
We carried out a literature review regarding the incidence, pathogenic mechanisms, risk factors, clinical presentation, and management strategies of KS-IRIS in the ART era.

2. Definition of KS-IRIS

General IRIS case definitions, according to French et al. and Shelburne et al., are reported in Table 1 [30,31]. The exaggerated and dysregulated cellular immune responses observed during IRIS could be due to an expansion of regulatory T cells that would not expand at the same rate as the antigen-specific effector cells [32]. Although this pathogen-specific immune response usually results in inflammation of tissues infected with the pathogen, cellular proliferative disease may also occur, explaining Hodgkin-lymphoma, non-Hodgkin lymphoma and KS-IRIS cases [8].
International consensus case definitions have been developed for tuberculosis-IRIS (TB-IRIS) [33] and cryptococcosis-IRIS [34], but not for KS-IRIS; however, there is a need for this [35]. This lack of international consensus case definitions might be explained, at least in part, by the lower incidence rate of KS-IRIS (see below in the incidence paragraph).
Therefore, definitions of KS-IRIS have varied according to different studies. French et al. defines KS-IRIS as either an abrupt clinical worsening of a previously existing KS or a new presentation of a previously unknown KS in temporal association with initiation or reinitiation of ART or change to a more active regimen. For each of these clinical situations, KS-IRIS occurrence is associated with a concomitant reduction of at least 1 log10 in the HIV-1 RNA level, or with two of the following three minor criteria: (a) a 2-fold increase in the CD4+ T-cell count after ART, (b) an increase in the immune response (KSHV-antibodies), and (c) a spontaneous resolution of disease without specific chemotherapy with continuation of ART [30].
In the study conducted by Volkow et al., KS-IRIS diagnosis was considered if a clinical abrupt worsening of previously existing KS (“paradoxical”) or the development of KS (“unmasked”) occurred within the first 6 months after the initiation of ART, associated with a reduction of at least 1 log10 of HIV-1 RNA and/or an increase of ≥50 cells/mm3 or ≥two fold rise in baseline CD4+ cell count. Furthermore, KS exacerbation was defined as having at least two of the following criteria: abrupt increase in the size or number of KS lesions, the appearance or exacerbation of lymphedema, and the appearance or increase of otherwise unexplained, gallium-negative, lung opacities on chest X-rays after starting ART [36].
Furthermore, in the absence of sufficient viral load reduction, Novak et al. proposed as an adequate response to ART a 50% or greater increase in CD4 cell count within 6 months after initiating a new or different ART regimen [2].
According to Bower et al., the major factors used to diagnose IRIS-associated KS are the temporal relationship between rapid clinical progression of KS and the initiation of ART ([28]). Thus, the lack of a standardized definition of KS-IRIS makes comparisons of incidence rates, risk factors, and the evaluation of therapeutic strategies of KS-IRIS difficult. In our opinion, the definition of KS-IRIS should combine the criteria proposed by Volkow et al. [36] and Novak et al. [2].
There is also a need to distinguish between KS-IRIS and a recurrence/occurrence of KS despite virological control under ART. We and others [16,17,32] have increasingly described cases of KS in virological controlled patients [37]. Most cases occur in well-controlled PLWH, under long-term antiretroviral therapy (and not at the start of ART), and do not usually correspond to the exacerbation of pre-existing Kaposi lesions. Several immunopathological hypotheses could explain such cases, including KSHV chronic antigen exposure, immune modulation by viral proteins, and local immune exhaustion, leading to the consideration of such KS presentation as a specific pattern of KS [22]. However, separating these two entities is essential, as their prognoses and therapeutic strategies differ.

3. Pathogenic Mechanisms of KS-IRIS

The pathogenesis of KS involves KSHV, cytokines, and the host-immune response.
In HIV-infected individuals, infection by KSHV is a necessary but not sufficient condition for KS development. Furthermore, not all HIV-KSHV coinfected individuals will develop the disease. Immunosuppression and HIV viral load have been found to be correlated with KS occurrence in PLWH [18,38]. Additionally, CD4:CD8 ratio <1 and CD8 hyperlymphocytosis greater than 1000/mm3 regardless of CD4 T-cell count have been found to be associated with an increased risk of KS [18,39].
HIV-1 infection might promote KSHV pathogenesis through the effects of HIV-1 proteins (tat protein) on KSHV replication and indirectly through CD4 lymphocyte depletion and the production of inflammatory cytokines (N-Kappa B activation, INF gamma, VEGF) [11,12,40].
KSHV induces viral and host cytokine production that promotes KS cell proliferation, differentiation and angiogenesis (IL-6, oncostatin, alfa-TNF, PDGF, VEGF) [10,41,42]. Previous studies have confirmed the major role of KSHV-specific T cellular immunity in the control of KS among PLWH [43,44,45,46,47].
Hence, successful control of KS obtained after ART initiation may be attributable to immune restoration and inhibition of HIV replication, thus decreasing HIV-1 angiogenic tat protein concentrations that promote KSHV survival and tumorigenesis within infected cells [48,49], and inhibiting intracellular cytokine production. The reduction of IL-6 limits the interaction between HIV-1 and KSHV and thus the progression of KS [50].
IRIS results from the rapid expansion of antigen-specific CD4+ and CD8+ lymphocytes following initiation of ART, with an imbalance between pro- and anti-inflammatory immune responses [51]. KS-IRIS may be partly explained by the restoration of a KSHV-specific immune T cell response that reduces KSHV replication while promoting paradoxical cellular proliferation and inflammation, leading to KS progression [51,52]. According to Letang et al., in patients with high KSHV dissemination and high HIV viral load, KSHV replication in tissue cells could not be controlled by the restored KSHV-specific immune responses, that could promotes a cytokine-induced reactive angioproliferation and tumorigenesis, resulting in the development of paradoxical KS-IRIS [35].

4. Incidence of KS-IRIS and Outcomes

A meta-analysis [53] of cohort studies published between 1998 and 2009 including 13,103 patients living with HIV starting ART reported 1699 (13%) cases of IRIS, of whom 6.4% were KS-IRIS. This rate appears to be much lower than the incidence rate of IRIS of 37.7% observed in patients with CMV retinitis [1].
The incidence of KS-IRIS varies widely, from 2.4% to 39%, depending on study design, populations, geographic regions, and the type of IRIS investigated (paradoxical vs. unmasking IRIS) [2,28,35,36,52,53,54,55,56,57]. The paradoxical KS-IRIS incidence in newly diagnosed HIV-positive patients initiating ART was reported to be 6% to11% in three studies from the USA, UK and Mozambique [28,53,54], and that of unmasking KS-IRIS in ART naïve patients, 5% and 4% in Mozambique [54] and a cohort in Mexico, respectively [36]. In two retrospective studies of patients living in the United States, the overall IRIS incidence rate in the first year of ART initiation or change was 11% [2,53]. However, in Achenbach et al.’s study investigating only paradoxical IRIS cases, the KS-IRIS incidence was 29% [53], while in a study performed by Novak et al., in which only unmasking IRIS cases were assessed, the KS-IRIS incidence was 2.4% [2]. Although there are few relevant studies, unmasking KS-IRIS seems to be less frequent compared to paradoxical KS-IRIS. However, some KS disease progression despite ART may have been misclassified as KS-IRIS [53].
KS-IRIS incidence and associated mortality were reported to be higher in low-resource settings compared to resource-rich countries [35]. In a pooled analysis of three prospective cohorts of ART-naïve HIV-infected patients with KS from sub-Saharan Africa and one from the UK, 13.9% of patients experienced paradoxical IRIS-KS, 20% among SSA cohorts and 8.5% in UK, with mortality 3.3-fold higher in the SSA cohorts. Factors predictive of mortality were KS-IRIS, lack of chemotherapy, pre-ART CD4 cell count of less than 200/mm3, and detectable baseline KSHV DNA in the plasma [35]. The prevalence of these factors was high in the African cohorts. Furthermore, the rate of KS-IRIS mortality observed in this study was higher than for any other paradoxical IRIS event associated with major opportunistic infections [35].
However, in two urban ART clinics in South Africa [55] with IRIS incidence 22.9%, of which 64% was unmasking-IRIS and 36% paradoxical IRIS, IRIS accounted for 24% of deaths, only one of which was due to paradoxical KS-IRIS [55].
The contribution to mortality of IRIS was also investigated among 2610 PLWH from the HOPS cohort in Washington. In this study, an increased risk of death was found in patients having experienced an IRIS event, with 23.2% of deaths versus 8.5% among those who had not, with a median delay of 40.6 months [2]. Unfortunately, risk of death and time delay to death was not assessed for each type of IRIS but visceral KS-IRIS was found to lead to considerable morbidity and mortality compared with IRIS related to other OIs [53].

5. KS-IRIS Associated Risk Factors

Most studies have examined risk factors for paradoxical and unmasking IRIS. and few for KS-IRIS [58,59].
In the study by Achenbach et al., a greater median increase in CD4 T cell count was identified in patients with mucocutaneous KS-IRIS [53]. A higher median increase in CD8 T cell count was also observed in patients with mucocutaneous KS who developed KS-IRIS. Such variations in CD4 and CD8 T cell count were not observed in patients with visceral KS who developed KS-IRIS [53].
Although low CD4 counts at the start of ART drive the incidence of IRIS independently of the pathology involved [1,36], KS-IRIS can occur at higher CD4 cell counts [28,60].
In a prospective cohort of 69 HIV infected adults conducted in Mozambique, Letang et al. identified four independent predictors of KS-IRIS including clinical pretreatment KS, detectable plasma KSHV DNA, hematocrit <30%, and high plasma HIV viral load (≥10 5 Log/mL) [52]. Additionally, in another study, T1 KS stage disease and high pre-ART plasma HIV VL were both reported to be associated with more than twice the risk of developing KS-IRIS, independently of the baseline CD4 cell counts, and KSHV DNA detectability in plasma was associated with a three-fold increase [35].
The association between T1 KS stage and KS-IRIS risk found in this study confirms the hypothesis that higher KS disease burden increases the risk of KS-IRIS, as suggested for IRIS associated with other OI [26,61,62,63]. The higher incidence of KS-IRIS observed in patients with visceral KS compared with patients with mucocutaneous KS reported in a study by Achenbach et al. confirmed these results [53]. Likewise, Volkow et al. reported that the extent and number of KS lesions as well as pulmonary involvement were significantly more in KS-IRIS individuals [36].
However, in a study by Bower et al., neither tumor stage nor site of KS were associated with KS-IRIS risk, while KS-associated edema at baseline was more likely to be observed in patients with KS-IRIS [28]. Likewise, edema interfering with physical function and raised cutaneous lesions before ART initiation were reported to be more frequent in patients who developed KS-IRIS [64].
Of note, concurrent or recent use of glucocorticoids may lead to increased risk of KS-IRIS [65].
Regarding KSHV DNA, detectable baseline plasma KSHV DNA was found to predict KS-IRIS. However, plasma KSHV DNA can be undetectable during KS-IRIS, as previously reported [66].

6. Clinical Presentations of KS-IRIS

The time of onset of KS-IRIS is variable, from a few days to 6 months after ART initiation, with the greatest risk within the first 2 months [4,27,28,35]. Haddow et al. have found that unmasking KS-IRIS occurred slightly but significantly later than paradoxical IRIS, with a median 21 days (IQR 7–52 days) and 11 days (IQR 7–24), respectively [55]. A longer median time of onset for mucocutaneous KS-IRIS cases than for other types of IRIS 144 vs. 36 days) has been reported by Achenbach et al., but the difference was not significant [53].
A short onset delay after ART initiation does not seem to have any predictive value, and KS-IRIS can remain a time-limited phenomenon, controlled with a limited course of early systemic chemotherapy [27] or continue to develop, despite several cycles of chemotherapy over a number of months [67].
Depending on the clinical presentation and the location of the lesions, KS-IRIS can be life-threatening but also lead to functional or aesthetic consequences.
Thus, paradoxical KS IRIS frequently presents as inflammation and enlargement of existing KS lesion with marked lesional swelling, increased tenderness, and peripheral edema or worsening edema [68]. It is of note that edema can persist after resolution of KS skin lesions [53].
KS may also extend or appear rapidly at new anatomical sites with variable symptomatology [58]. In its disseminated form, lymph node and visceral involvement can be observed, in particular in the respiratory and gastrointestinal tract, but every organ can potentially be affected, such as bone and cartilage [69] or kidney [70]. In case of gastrointestinal tract involvement, although most often asymptomatic, significant complications may occur, including perforation, bleeding and obstruction [71,72].
Pulmonary manifestations include cough, dyspnea, hemoptysis, parenchymal nodular lesions, adenopathy, and pleural effusions that can necessitate thoracentesis and talc pleurodesis [73]. The presence of endobronchial lesions can lead to acute airway obstruction, which is potentially life-threatening [51,74].
More atypical presentations of KS-IRIS have been reported, such as oral localization [75], chylous ascites and chylothorax [67]. Although rare, KS-IRIS can also be observed in children, as recently reported in a 9-year-old Guinean girl [76].
Diagnosis of KS can be confirmed with immunohistochemical staining using endothelial markers such as CD31, CD34, D2-40, and ERG, through skin biopsy or alternatively endoscopic, pleural ortransbronchial biopsy. No current guidelines recommend the use of virological diagnosis tools to detect the HHV-8 genome or antibodies for KS diagnosis, but HHV-8 can be identified by using LNA-1 antibody or by PCR. Imaging using modalities such as CT, MRI, and FDG-PET/CT scans can aid in diagnosing and staging cutaneous KS and lymph node involvement, as well as responses to treatment of KS lesions [77,78].
Both paradoxical and unmasking KS-IRIS have been associated with significant morbidity and mortality in patients with visceral disease, including persistence of mucocutaneous lesions, lymphedema and death [26,27,79,80,81], sometimes occurring shortly after IRIS diagnosis [53]. Pulmonary involvement is one of the major risks for mortality, with a prevalence reported to be 19 to 35%. [27,36,79,82]. Thrombocytopenia (<100,000 platelets/mm3 at 12 weeks) was found to be significantly associated with death [36].
More severe clinical presentations have been reported, with abrupt clinical exacerbation, fever, thrombocytopenia, anemia, hyponatremia, and hypoalbuminemia [83]. However, these symptoms combined with signs of systemic inflammatory syndrome (SIRS) whould suggest the diagnosis of Kaposi Sarcoma Inflammatory Cytokine Syndrome (KICS).This syndrome is associated with a high rate of morbidity and mortality, and its diagnosis requires excisional biopsy of lymphadenopathy to exclude multicentric Castleman disease [84] and facilitate the initiation of prompt treatment [85,86].

7. Prevention of KS-IRIS

In contrast with the prevention of TB-IRIS which relies on a delay of ART initiation of at least four weeks after TB treatment initiation [87], ART should not to be delayed in the case of KS. The controversy regarding IRIS risk after ART initiation including integrase inhibitors [88,89], appears to be unfounded, following the results of a recent meta-analysis [90].
The best therapeutic approach for the prevention of KS-IRIS is still not currently defined. Management of AIDS-related Kaposi sarcoma depends on the location and extent of the disease, according to most guidelines [68,91]. Indications for systemic therapy include visceral disease, painful or ulcerated lesions, edema, extensive cutaneous disease, rapidly progressive disease, psychological withdrawal because of the stigma of visible lesions, and KS-IRIS [92]. In other cases, maintaining ART without additional systemic chemotherapy may be sufficient.
Thus, decisions about whether to initiate chemotherapy together with ART to prevent KS-IRIS must consider the extent of the diseaseand the potential benefits and added toxicities of chemotherapy plus ART over ART alone, in particular for individuals with severe immunosuppression. Furthermore, potential drug–drug interactions between ART and chemotherapy need to be assessed [93]. Whatever the decision, close monitoring of patients should be implemented in order to identify the first manifestations of KS-IRIS as quickly as possible.

7.1. Chemotherapy for Reduction of KS-IRIS Risk

A meta-analysis of six randomized trials and three observational studies involving 792 HIV-infected patients with severe KS has shown that ART plus chemotherapy with paclitaxel, liposomal doxorubicin, or daunorubicin facilitated a significant reduction of disease progression, with no differences between the three drugs [94], but no statistically significant reduction in KS-IRIS, as had been observed in one UK cohort study [95]. In this cohort, liposomal anthracyclines plus ART were compared to ART alone in 129 patients with T1KS, with the following results: four out of 65 T1 participants in the ART plus liposomal anthracycline group developed KS- IRIS, compared to eight out of 64 participants in the ART only group (RR 0.49; 95% CI 0.16 to 1.55) [95].
However, Letang et al. reported, in a pooled analysis of three prospective cohorts of ART-naïve PLWH with KS from SAA and one from the UK, a positive impact of adjunctive chemotherapy on KS-IRIS risk [35]. This result was consistent with previous studies conducted in South Africa and Zimbabwe, showing better control of KSHV replication and clinical outcomes among KS patients receiving ART and chemotherapy, vs. ART only [43,96,97].
Recently, three randomized clinical trials conducted in SSA and Brazil evaluated different chemotherapy drug regimens for the prevention of KS-IRIS. However, liposomal anthracyclines, the gold standard of care, are not available in these countries, and only non-liposomal doxorubicin, bleomycin, vincristine, and etoposide may be currently administered, even though they are less effective and have worse side-effect profiles in cases of KS [92].
A randomized trial conducted by Mosam et al. in South Africa found that the addition of chemotherapy with intravenous bleomycin/vincristine to ART did not affect rates of KS-IRIS [96]. In a three-arm randomized clinical trial conducted by Krown et al. in SSA countries and Brazil, suspected KS-IRIS occurred infrequently during treatment of advanced and symptomatic KS with ART in combination with each of three chemotherapy regimens tested, as follows: 10% in etoposide, 2% in bleomycin/vincristine, and none in the paclitaxel arm [98].
In a randomized trial evaluating the impact of initiating ART with either immediate or as-needed oral etoposide in PLWH with mild to moderate KS, immediate etoposide with ART provided early but non-durable clinical benefits [99], and reduced the incidence of KS-IRIS, as follows: 7% in the immediate etoposide with ART arm, versus 21% in the ART only arm [64]. In this trial, etoposide was administered as a single 50mg capsule on days 1–7 of each 14 day cycle.

7.2. Antiviral Agent in Reduction of KS-IRIS Risk

Valganciclovir has been evaluated in the prevention of KS-IRIS due to the in vitro activity of this antiviral agent against KSHV and epidemiological data showing a reduction of KS incidence when given prophylactically for CMV infection [100,101,102,103,104,105]. The randomized clinical trial conducted by Volkow et al. on 38 ART naïve patients with disseminated KS found a reduction of severe KS-IRIS events and in the number of patients with at least one severe KS-IRIS episode. Valganciclovir treatment was administered at 900 mg BID, initiated four weeks before ART and maintained for 48 weeks [106]. This result remains to be confirmed in a larger number of patients, and the benefits and risks of such a prophylaxis weighed, particularly due to valganciclovir’s haematological toxicities.

8. Management of KS-IRIS

KS-IRIS does not indicate the failure of ART or a need for changes in ART regimen [43,58], and systemic chemotherapy for KS in conjunction with ART is recommended [92].
The staging for AIDS-related KS was developed in adults by the AIDS Clinical Trial Group (ACTG) of the National Institutes of Health in the pre-ART era [107] and was revised in 1997 [108]. This staging is based on the extent of the tumor, immune status, and severity of systemic illness (Table 2) (TIS classification). This classification is more often used in research than in clinical practice, where patients are grouped into (1) clinically severe KS needing an immediate fast-acting intervention, or (2) clinically mild to moderate KS, for which patients can be treated with ART alone. However, the definition of severe KS remains debatable, but disease progression can be evaluated according to the ACTG response criteria (Table 3) [107].
Currently, liposomal anthracyclines and paclitaxel are the two reference treatment for patients suffering from extensive and/or rapidly progressive KS, and liposomal anthracyclines are the gold standard of care. However, to our knowledge, the best therapeutic approach for KS-IRIS has not yet yet evaluated in controlled trials.
In contrast with management of IRIS for some opportunistic infections, glucocorticoids may be harmful for KS-IRIS and are contra-indicated, because of the potential for life-threatening KS exacerbation [65,74,109,110]. Therefore, administration of glucocorticoids should be limited to life-threatening conditions for which glucocorticoids are otherwise indicated (i.e., anaphylaxis) [68], on a case-by-case basis after an assessment of risks and benefits, as in a case report [66], and under close monitoring for the early detection of KS lesions.
In recent years, multiple targeted therapies for KS have been evaluated in clinical trials but, to our knowledge, none in patients with KS-IRIS [111,112,113,114,115,116,117,118,119], which means we are unable to define their place in the prevention or treatment of KS-IRIS. Among these targeted therapies, the demonstration of PD-1 expression in KS has led to the evaluation of immune-checkpoint-inhibitors (ICI) as a possible treatment option for KS [120,121]. A systematic review of the safety and efficacy of ICI therapy in HIV-infected patients with advanced-stage cancer reported an incidence of 8.6% of grade 3 or higher immune-related adverse events (irAEs) [122] but, to our knowledge, no cases of IRIS.
Although no cases of irAEs related to anti-PD-1 therapy have been observed in a group of [9] HIV positive patients with KS 119, clinicians should bear in mind that steroids, which are generally used for irAEs [123], should be avoided in patients with active or prior KS, given the potential to cause significant flares and relapses [68]. Likewise, other therapies that suppress B-and T-cell numbers and /or function, such as rituximab are associated with flares of KS and should be used with caution [124].
Moreover, a case of KSHV-associated B-cell lymphoproliferation, possibly attributable to pembrolizumab, has been observed in a heavily pretreated patient with KS and a history of elevated peripheral circulating cell-associated KSHV and KICS; this emphasizes the need for careful monitoring of these patients during treatment with targeted therapies [125].

9. Conclusions

Although KS-IRIS is less frequent than other IRIS-related opportunistic infections, close clinical supervision is warranted for patients with KS who are initiating, changing, or resuming ART, as the onset of IRIS-associated KS flare can be observed in as little as three weeks. Particular vigilance is recommended in the case of KS with visceral involvement and peripheral edema, including in those with minimally altered immune status. Clinicians should remain cautious as IRIS-KS is often not readily recognized and may occur in patients resuming ART after a long interruption period. No preventive treatment for KS-IRIS has yet been validated, and glucocorticoids may be harmful for KS-IRIS treatment, therefore they should be limited to life-threatening cases only.

Author Contributions

Writing—original draft preparation, I.P.-M.; writing—review and editing, I.P.-M., A.M., R.P.; visualization, S.B., A.-G.M., C.S., M.V., M.-A.V., J.-P.S.; supervision, J.-P.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Teresa Rojas Rojas and Yamile Brouri for administrative support.

Conflicts of Interest

Isabelle Poizot-Martin: consultancies (Gilead Sciences, MSD), travel/accommodation/meeting expenses (Gilead sciences, ViiVhealthcare), outside the submitted work. Sylvie Brégigeon: congress grants (ViiV, Healthcare), consultancies (ViiV Healthcare, Gilead, Janssen, MSD), outside the submitted work. Romain Palich has declared no conflict of interest with the submitted work. Anne-Geneviève Marcelin: consultancies (Gilead Sciences, VIIV Healthcare, MSD, Theratechnologies), research grants (Gilead sciences, ViiV Healthcare, MSD), outside the submitted work. Marc-Antoine Valantin has declared no conflict of interest with the submitted work. Caroline Solas has declared no conflict of interest with the submitted work. Marianne Veyri has declared no conflict of interest with the submitted work. Jean-Philippe Spano has no direct conflict of interest but received an honorarium as a consultant for Roche, MSD and Biogaran and has received a speaker honorarium from MSD, Roche, AstraZeneca, Leopharma, Mylan, Pfizer, BMS, Novartis, PFO, Myriads, Gilead and Lilly. Alain Makinson: travel grants (MSD, Mylan; Donations (MSD), outside the submitted work. The other authors declare no conflict of interest.

References

  1. Müller, M.; Wandel, S.; Colebunders, R.; Attia, S.; Furrer, H.; Egger, M. IeDEA Southern and Central Africa Immune reconstitution inflammatory syndrome in patients starting antiretroviral therapy for HIV infection: A systematic review and meta-analysis. Lancet Infect. Dis. 2010, 10, 251–261. [Google Scholar] [CrossRef] [Green Version]
  2. Novak, R.M.; Richardson, J.T.; Buchacz, K.; Chmiel, J.S.; Durham, M.D.; Palella, F.J.; Wendrow, A.; Wood, K.; Young, B.; Brooks, J.T.; et al. Immune reconstitution inflammatory syndrome: Incidence and implications for mortality. AIDS 2012, 26, 721–730. [Google Scholar] [CrossRef] [Green Version]
  3. Sereti, I.; Rodger, A.J.; French, M.A. Biomarkers in immune reconstitution inflammatory syndrome: Signals from pathogenesis. Curr. Opin. HIV AIDS 2010, 5, 504–510. [Google Scholar] [CrossRef] [Green Version]
  4. Feller, L.; Lemmer, J. Insights into pathogenic events of HIV-associated Kaposi sarcoma and immune reconstitution syndrome related Kaposi sarcoma. Infect. Agent Cancer 2008, 3, 1–9. [Google Scholar] [CrossRef] [Green Version]
  5. Ghosn, J.; Taiwo, B.; Seedat, S.; Autran, B.; Katlama, C. HIV. Lancet 2018, 392, 685–697. [Google Scholar] [CrossRef]
  6. French, M.A.; Lenzo, N.; John, M.; Mallal, S.A.; McKinnon, E.J.; James, I.R.; Price, P.; Flexman, J.P.; Tay-Kearney, M.L. Immune restoration disease after the treatment of immunodeficient HIV-infected patients with highly active antiretroviral therapy. HIV Med. 2000, 1, 107–115. [Google Scholar] [CrossRef]
  7. Cattelan, A.M.; Mattiolo, A.; Grassi, A.; Piano, M.A.; Sasset, L.; Trevenzoli, M.; Zanovello, P.; Calabrò, M.L. Predictors of immune reconstitution inflammatory syndrome associated with Kaposi’s sarcoma: A case report. Infect. Agent Cancer 2016, 11, 1–4. [Google Scholar] [CrossRef] [Green Version]
  8. Vishnu, P.; Dorer, R.P.; Aboulafia, D.M. Immune Reconstitution Inflammatory Syndrome-Associated Burkitt Lymphoma After Combination Antiretroviral Therapy in HIV-Infected Patients. Clin. Lymphoma Myeloma Leuk. 2015, 15, e23–e29. [Google Scholar] [CrossRef] [Green Version]
  9. Gopal, S.; Patel, M.R.; Yanik, E.L.; Cole, S.R.; Achenbach, C.J.; Napravnik, S.; Burkholder, G.A.; Reid, E.G.; Rodriguez, B.; Deeks, S.G.; et al. Temporal trends in presentation and survival for HIV-associated lymphoma in the antiretroviral therapy era. J. Natl. Cancer Inst. 2013, 105, 1221–1229. [Google Scholar] [CrossRef]
  10. Chang, Y.; Cesarman, E.; Pessin, M.S.; Lee, F.; Culpepper, J.; Knowles, D.M.; Moore, P.S. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 1994, 266, 1865–1869. [Google Scholar] [CrossRef] [Green Version]
  11. Nakamura, S.; Salahuddin, S.Z.; Biberfeld, P.; Ensoli, B.; Markham, P.D.; Wong-Staal, F.; Gallo, R.C. Kaposi’s sarcoma cells: Long-term culture with growth factor from retrovirus-infected CD4+ T cells. Science 1988, 242, 426–430. [Google Scholar] [CrossRef] [PubMed]
  12. Ensoli, B.; Barillari, G.; Salahuddin, S.Z.; Gallo, R.C.; Wong-Staal, F. Tat protein of HIV-1 stimulates growth of cells derived from Kaposi’s sarcoma lesions of AIDS patients. Nature 1990, 345, 84–86. [Google Scholar] [CrossRef]
  13. Boshoff, C.; Chang, Y. Kaposi’s sarcoma-associated herpesvirus: A new DNA tumor virus. Annu. Rev. Med. 2001, 52, 453–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Mesri, E.A.; Feitelson, M.A.; Munger, K. Human viral oncogenesis: A cancer hallmarks analysis. Cell Host Microbe 2014, 15, 266–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Jha, H.C.; Banerjee, S.; Robertson, E.S. The Role of Gammaherpesviruses in Cancer Pathogenesis. Pathogens 2016, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  16. La Ferla, L.; Pinzone, M.R.; Nunnari, G.; Martellotta, F.; Lleshi, A.; Tirelli, U.; De Paoli, P.; Berretta, M.; Cacopardo, B. Kaposi’ s sarcoma in HIV-positive patients: The state of art in the HAART-era. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 2354–2365. [Google Scholar]
  17. Bohlius, J.; Valeri, F.; Maskew, M.; Prozesky, H.; Garone, D.; Sengayi, M.; Fox, M.P.; Davies, M.-A.; Egger, M. Kaposi’s Sarcoma in HIV-infected patients in South Africa: Multicohort study in the antiretroviral therapy era. Int. J. Cancer 2014, 135, 2644–2652. [Google Scholar] [CrossRef] [Green Version]
  18. Poizot-Martin, I.; Lions, C.; Cheret, A.; Rey, D.; Duvivier, C.; Jacomet, C.; Allavena, C.; Huleux, T.; Bani-Sadr, F.; Obry-Roguet, V.; et al. Kaposi sarcoma in people living with HIV: Incidence and associated factors in a French cohort between 2010 and 2015. AIDS 2020, 34, 569–577. [Google Scholar] [CrossRef]
  19. Hernández-Ramírez, R.U.; Shiels, M.S.; Dubrow, R.; Engels, E.A. Cancer risk in HIV-infected people in the USA from 1996 to 2012: A population-based, registry-linkage study. Lancet HIV 2017, 4, e495–e504. [Google Scholar] [CrossRef]
  20. Poizot-Martin, I.; Lions, C.; Allavena, C.; Huleux, T.; Bani-Sadr, F.; Cheret, A.; Rey, D.; Duvivier, C.; Jacomet, C.; Ferry, T.; et al. Spectrum and Incidence Trends of AIDS- and Non-AIDS-Defining Cancers between 2010 and 2015 in the French Dat’AIDS Cohort. Cancer Epidemiol. Biomark. Prev. 2021, 30, 554–563. [Google Scholar] [CrossRef]
  21. Yanik, E.L.; Napravnik, S.; Cole, S.R.; Achenbach, C.J.; Gopal, S.; Olshan, A.; Dittmer, D.P.; Kitahata, M.M.; Mugavero, M.J.; Saag, M.; et al. Incidence and timing of cancer in HIV-infected individuals following initiation of combination antiretroviral therapy. Clin. Infect. Dis. 2013, 57, 756–764. [Google Scholar] [CrossRef] [Green Version]
  22. Palich, R.; Veyri, M.; Valantin, M.-A.; Marcelin, A.-G.; Guihot, A.; Pourcher, V.; Jary, A.; Solas, C.; Makinson, A.; Poizot-Martin, I.; et al. Recurrence and Occurrence of Kaposi’s Sarcoma in Patients Living With Human Immunodeficiency Virus (HIV) and on Antiretroviral Therapy, Despite Suppressed HIV Viremia. Clin. Infect. Dis. 2020, 70, 2435–2438. [Google Scholar] [CrossRef]
  23. Séverin, D.; Bessaoud, F.; Meftah, N.; Du Thanh, A.; Tretarre, B.; Guillot, B.; Makinson, A. A comparative study of classic and HIV-viremic and aviremic AIDS Kaposi sarcoma. AIDS 2021, 35, 399–405. [Google Scholar] [CrossRef] [PubMed]
  24. Poizot-Martin, I.; Obry-Roguet, V.; Duvivier, C.; Lions, C.; Huleux, T.; Jacomet, C.; Ferry, T.; Cheret, A.; Allavena, C.; Bani-Sadr, F.; et al. Kaposi sarcoma among people living with HIV in the French DAT’AIDS cohort between 2010 and 2015. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 1065–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Engels, E.A.; Yanik, E.L.; Wheeler, W.; Gill, M.J.; Shiels, M.S.; Dubrow, R.; Althoff, K.N.; Silverberg, M.J.; Brooks, J.T.; Kitahata, M.M.; et al. Cancer-Attributable Mortality Among People With Treated Human Immunodeficiency Virus Infection in North America. Clin. Infect. Dis. 2017, 65, 636–643. [Google Scholar] [CrossRef] [PubMed]
  26. Connick, E.; Kane, M.A.; White, I.E.; Ryder, J.; Campbell, T.B. Immune reconstitution inflammatory syndrome associated with Kaposi sarcoma during potent antiretroviral therapy. Clin. Infect. Dis. 2004, 39, 1852–1855. [Google Scholar] [CrossRef] [PubMed]
  27. Leidner, R.S.; Aboulafia, D.M. Recrudescent Kaposi’s sarcoma after initiation of HAART: A manifestation of immune reconstitution syndrome. AIDS Patient Care STDS 2005, 19, 635–644. [Google Scholar] [CrossRef]
  28. Bower, M.; Nelson, M.; Young, A.M.; Thirlwell, C.; Newsom-Davis, T.; Mandalia, S.; Dhillon, T.; Holmes, P.; Gazzard, B.G.; Stebbing, J. Immune reconstitution inflammatory syndrome associated with Kaposi’s sarcoma. J. Clin. Oncol. 2005, 23, 5224–5228. [Google Scholar] [CrossRef]
  29. Jaffe, H.W.; De Stavola, B.L.; Carpenter, L.M.; Porter, K.; Cox, D.R. CASCADE Collaboration Immune reconstitution and risk of Kaposi sarcoma and non-Hodgkin lymphoma in HIV-infected adults. AIDS 2011, 25, 1395–1403. [Google Scholar] [CrossRef]
  30. French, M.A.; Price, P.; Stone, S.F. Immune restoration disease after antiretroviral therapy. AIDS 2004, 18, 1615–1627. [Google Scholar] [CrossRef]
  31. Shelburne, S.A.; Montes, M.; Hamill, R.J. Immune reconstitution inflammatory syndrome: More answers, more questions. J. Antimicrob. Chemother. 2006, 57, 167–170. [Google Scholar] [CrossRef] [PubMed]
  32. Ruhwald, M.; Ravn, P. Immune reconstitution syndrome in tuberculosis and HIV-co-infected patients: Th1 explosion or cytokine storm? AIDS 2007, 21, 882–884. [Google Scholar] [CrossRef]
  33. Meintjes, G.; Lawn, S.D.; Scano, F.; Maartens, G.; French, M.A.; Worodria, W.; Elliott, J.H.; Murdoch, D.; Wilkinson, R.J.; Seyler, C.; et al. Tuberculosis-associated immune reconstitution inflammatory syndrome: Case definitions for use in resource-limited settings. Lancet Infect. Dis. 2008, 8, 516–523. [Google Scholar] [CrossRef] [Green Version]
  34. Haddow, L.J.; Colebunders, R.; Meintjes, G.; Lawn, S.D.; Elliott, J.H.; Manabe, Y.C.; Bohjanen, P.R.; Sungkanuparph, S.; Easterbrook, P.J.; French, M.A.; et al. Cryptococcal immune reconstitution inflammatory syndrome in HIV-1-infected individuals: Proposed clinical case definitions. Lancet Infect. Dis. 2010, 10, 791–802. [Google Scholar] [CrossRef] [Green Version]
  35. Letang, E.; Lewis, J.J.; Bower, M.; Mosam, A.; Borok, M.; Campbell, T.B.; Naniche, D.; Newsom-Davis, T.; Shaik, F.; Fiorillo, S.; et al. Immune reconstitution inflammatory syndrome associated with Kaposi sarcoma: Higher incidence and mortality in Africa than in the UK. AIDS 2013, 27, 1603–1613. [Google Scholar] [CrossRef] [PubMed]
  36. Volkow, P.; Cesarman-Maus, G.; Garciadiego-Fossas, P.; Rojas-Marin, E.; Cornejo-Juárez, P. Clinical characteristics, predictors of immune reconstitution inflammatory syndrome and long-term prognosis in patients with Kaposi sarcoma. AIDS Res. Ther. 2017, 14, 30. [Google Scholar] [CrossRef] [PubMed]
  37. Khanlou, H.; Stein, T.; Farthing, C. Development of Kaposi sarcoma despite sustained suppression of HIV plasma viremia. J. Acquir. Immune Defic. Syndr. 2000, 23, 361. [Google Scholar] [CrossRef]
  38. Dubrow, R.; Qin, L.; Lin, H.; Hernández-Ramírez, R.U.; Neugebauer, R.S.; Leyden, W.; Althoff, K.N.; Achenbach, C.J.; Hessol, N.A.; Modur, S.P.; et al. Association of CD4+ T-cell Count, HIV-1 RNA Viral Load, and Antiretroviral Therapy With Kaposi Sarcoma Risk Among HIV-infected Persons in the United States and Canada. J. Acquir. Immune Defic. Syndr. 2017, 75, 382–390. [Google Scholar] [CrossRef]
  39. Caby, F.; Guiguet, M.; Weiss, L.; Winston, A.; Miro, J.M.; Konopnicki, D.; Le Moing, V.; Bonnet, F.; Reiss, P.; Mussini, C.; et al. CD4/CD8 Ratio and the Risk of Kaposi Sarcoma or Non-Hodgkin Lymphoma in the Context of Efficiently Treated Human Immunodeficiency Virus (HIV) Infection: A Collaborative Analysis of 20 European Cohort Studies. Clin. Infect. Dis. 2021, 73, 50–59. [Google Scholar] [CrossRef]
  40. Varthakavi, V.; Browning, P.J.; Spearman, P. Human immunodeficiency virus replication in a primary effusion lymphoma cell line stimulates lytic-phase replication of Kaposi’s sarcoma-associated herpesvirus. J. Virol. 1999, 73, 10329–10338. [Google Scholar] [CrossRef] [Green Version]
  41. Hengge, U.R.; Ruzicka, T.; Tyring, S.K.; Stuschke, M.; Roggendorf, M.; Schwartz, R.A.; Seeber, S. Update on Kaposi’s sarcoma and other HHV8 associated diseases. Part 1: Epidemiology, environmental predispositions, clinical manifestations, and therapy. Lancet Infect. Dis. 2002, 2, 281–292. [Google Scholar] [CrossRef]
  42. Gantt, S.; Casper, C. Human herpesvirus 8-associated neoplasms: The roles of viral replication and antiviral treatment. Curr. Opin. Infect. Dis. 2011, 24, 295–301. [Google Scholar] [CrossRef] [Green Version]
  43. Bihl, F.; Mosam, A.; Henry, L.N.; Chisholm, J.V.; Dollard, S.; Gumbi, P.; Cassol, E.; Page, T.; Mueller, N.; Kiepiela, P.; et al. Kaposi’s sarcoma-associated herpesvirus-specific immune reconstitution and antiviral effect of combined HAART/chemotherapy in HIV clade C-infected individuals with Kaposi’s sarcoma. AIDS 2007, 21, 1245–1252. [Google Scholar] [CrossRef]
  44. Bihl, F.; Berger, C.; Chisholm, J.V.; Henry, L.M.; Bertisch, B.; Trojan, A.; Nadal, D.; Speck, R.F.; Flepp, M.; Brander, C.; et al. Cellular immune responses and disease control in acute AIDS-associated Kaposi’s sarcoma. AIDS 2009, 23, 1918–1922. [Google Scholar] [CrossRef] [Green Version]
  45. Bourboulia, D.; Aldam, D.; Lagos, D.; Allen, E.; Williams, I.; Cornforth, D.; Copas, A.; Boshoff, C. Short- and long-term effects of highly active antiretroviral therapy on Kaposi sarcoma-associated herpesvirus immune responses and viraemia. AIDS 2004, 18, 485–493. [Google Scholar] [CrossRef]
  46. Guihot, A.; Dupin, N.; Marcelin, A.-G.; Gorin, I.; Bedin, A.-S.; Bossi, P.; Galicier, L.; Oksenhendler, E.; Autran, B.; Carcelain, G. Low T cell responses to human herpesvirus 8 in patients with AIDS-related and classic Kaposi sarcoma. J. Infect. Dis. 2006, 194, 1078–1088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Lambert, M.; Gannagé, M.; Karras, A.; Abel, M.; Legendre, C.; Kerob, D.; Agbalika, F.; Girard, P.-M.; Lebbe, C.; Caillat-Zucman, S. Differences in the frequency and function of HHV8-specific CD8 T cells between asymptomatic HHV8 infection and Kaposi sarcoma. Blood 2006, 108, 3871–3880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Zhou, F.; Xue, M.; Qin, D.; Zhu, X.; Wang, C.; Zhu, J.; Hao, T.; Cheng, L.; Chen, X.; Bai, Z.; et al. HIV-1 Tat promotes Kaposi’s sarcoma-associated herpesvirus (KSHV) vIL-6-induced angiogenesis and tumorigenesis by regulating PI3K/PTEN/AKT/GSK-3β signaling pathway. PLoS ONE 2013, 8, e53145. [Google Scholar] [CrossRef] [Green Version]
  49. Pati, S.; Foulke, J.S.; Barabitskaya, O.; Kim, J.; Nair, B.C.; Hone, D.; Smart, J.; Feldman, R.A.; Reitz, M. Human herpesvirus 8-encoded vGPCR activates nuclear factor of activated T cells and collaborates with human immunodeficiency virus type 1 Tat. J. Virol. 2003, 77, 5759–5773. [Google Scholar] [CrossRef] [Green Version]
  50. Stebbing, J.; Portsmouth, S.; Gazzard, B. How does HAART lead to the resolution of Kaposi’s sarcoma? J. Antimicrob. Chemother. 2003, 51, 1095–1098. [Google Scholar] [CrossRef] [Green Version]
  51. Stover, K.R.; Molitorisz, S.; Swiatlo, E.; Muzny, C.A. A fatal case of Kaposi sarcoma due to immune reconstitution inflammatory syndrome. Am. J. Med. Sci. 2012, 343, 421–425. [Google Scholar] [CrossRef] [PubMed]
  52. Letang, E.; Almeida, J.M.; Miró, J.M.; Ayala, E.; White, I.E.; Carrilho, C.; Bastos, R.; Nhampossa, T.; Menéndez, C.; Campbell, T.B.; et al. Predictors of immune reconstitution inflammatory syndrome-associated with Kaposi sarcoma in mozambique: A prospective study. J. Acquir. Immune Defic. Syndr. 2010, 53, 589–597. [Google Scholar] [CrossRef] [PubMed]
  53. Achenbach, C.J.; Harrington, R.D.; Dhanireddy, S.; Crane, H.M.; Casper, C.; Kitahata, M.M. Paradoxical Immune Reconstitution Inflammatory Syndrome in HIV-Infected Patients Treated With Combination Antiretroviral Therapy After AIDS-Defining Opportunistic Infection. Clin. Infect. Dis. 2012, 54, 424–433. [Google Scholar] [CrossRef] [PubMed]
  54. Letang, E.; Miró, J.M.; Nhampossa, T.; Ayala, E.; Gascon, J.; Menéndez, C.; Alonso, P.L.; Naniche, D. Incidence and predictors of immune reconstitution inflammatory syndrome in a rural area of Mozambique. PLoS ONE 2011, 6, e16946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Haddow, L.J.; Moosa, M.-Y.S.; Mosam, A.; Moodley, P.; Parboosing, R.; Easterbrook, P.J. Incidence, Clinical Spectrum, Risk Factors and Impact of HIV-Associated Immune Reconstitution Inflammatory Syndrome in South Africa. PLoS ONE 2012, 7, e40623. [Google Scholar] [CrossRef]
  56. Hoyo-Ulloa, I.; Belaunzarán-Zamudio, P.F.; Crabtree-Ramirez, B.; Galindo-Fraga, A.; Pérez-Aguinaga, M.E.; Sierra-Madero, J.G. Impact of the immune reconstitution inflammatory syndrome (IRIS) on mortality and morbidity in HIV-infected patients in Mexico. Int. J. Infect. Dis. 2011, 15, e408–e414. [Google Scholar] [CrossRef] [Green Version]
  57. Kumar, S.R.; Gopalan, N.; Patrawalla, P.; Menon, P.; Mayer, K.; Swaminathan, S. Immune reconstitution inflammatory syndrome in HIV-infected patients with and without prior tuberculosis. Int. J. STD AIDS 2012, 23, 419–423. [Google Scholar] [CrossRef] [Green Version]
  58. Walker, N.F.; Scriven, J.; Meintjes, G.; Wilkinson, R.J. Immune reconstitution inflammatory syndrome in HIV-infected patients. HIV AIDS 2015, 7, 49–64. [Google Scholar] [CrossRef] [Green Version]
  59. Vinhaes, C.L.; Araujo-Pereira, M.; Tibúrcio, R.; Cubillos-Angulo, J.M.; Demitto, F.O.; Akrami, K.M.; Andrade, B.B. Systemic Inflammation Associated with Immune Reconstitution Inflammatory Syndrome in Persons Living with HIV. Life 2021, 11, 65. [Google Scholar] [CrossRef]
  60. Mocroft, A.; Youle, M.; Phillips, A.N.; Halai, R.; Easterbrook, P.; Johnson, M.A.; Gazzard, B. The incidence of AIDS-defining illnesses in 4883 patients with human immunodeficiency virus infection. Royal Free/Chelsea and Westminster Hospitals Collaborative Group. Arch. Intern. Med. 1998, 158, 491–497. [Google Scholar] [CrossRef] [Green Version]
  61. Lortholary, O.; Fontanet, A.; Mémain, N.; Martin, A.; Sitbon, K.; Dromer, F. French Cryptococcosis Study Group Incidence and risk factors of immune reconstitution inflammatory syndrome complicating HIV-associated cryptococcosis in France. AIDS 2005, 19, 1043–1049. [Google Scholar] [CrossRef] [PubMed]
  62. Robertson, J.; Meier, M.; Wall, J.; Ying, J.; Fichtenbaum, C.J. Immune reconstitution syndrome in HIV: Validating a case definition and identifying clinical predictors in persons initiating antiretroviral therapy. Clin. Infect. Dis. 2006, 42, 1639–1646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sungkanuparph, S.; Filler, S.G.; Chetchotisakd, P.; Pappas, P.G.; Nolen, T.L.; Manosuthi, W.; Anekthananon, T.; Morris, M.I.; Supparatpinyo, K.; Kopetskie, H.; et al. Cryptococcal immune reconstitution inflammatory syndrome after antiretroviral therapy in AIDS patients with cryptococcal meningitis: A prospective multicenter study. Clin. Infect. Dis. 2009, 49, 931–934. [Google Scholar] [CrossRef]
  64. Nyirenda, M.; Ngongondo, M.; Kang, M.; Umbleja, T.; Krown, S.E.; Godfrey, C.; Samaneka, W.; Mngqibisa, R.; Hoagland, B.; Mwelase, N.; et al. Early Progression and Immune Reconstitution Inflammatory Syndrome During Treatment of Mild-To-Moderate Kaposi Sarcoma in Sub-Saharan Africa and South America: Incidence, Long-Term Outcomes, and Effects of Early Chemotherapy. J. Acquir. Immune Defic. Syndr. 2020, 84, 422–429. [Google Scholar] [CrossRef] [PubMed]
  65. Fernández-Sánchez, M.; Iglesias, M.C.; Ablanedo-Terrazas, Y.; Ormsby, C.E.; Alvarado-de la Barrera, C.; Reyes-Terán, G. Steroids are a risk factor for Kaposi’s sarcoma-immune reconstitution inflammatory syndrome and mortality in HIV infection. AIDS 2016, 30, 909–914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Speicher, D.J.; Sehu, M.M.; Johnson, N.W.; Shaw, D.R. Successful treatment of an HIV-positive patient with unmasking Kaposi’s sarcoma immune reconstitution inflammatory syndrome. J. Clin. Virol. 2013, 57, 282–285. [Google Scholar] [CrossRef]
  67. Cipriano, P.; Nabais, I.; Melo, N.; Alves, A.R. Chylous ascites in disseminated Kaposi sarcoma: An unusual manifestation as immune reconstitution inflammatory syndrome. BMJ Case Rep. 2019, 12, e228406. [Google Scholar] [CrossRef]
  68. Reid, E.; Suneja, G.; Ambinder, R.F.; Ard, K.; Baiocchi, R.; Barta, S.K.; Carchman, E.; Cohen, A.; Crysler, O.V.; Gupta, N.; et al. AIDS-Related Kaposi Sarcoma, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2019, 17, 171–189. [Google Scholar] [CrossRef] [Green Version]
  69. Lu, C.-L.; Chang, S.-Y.; Tseng, Y.-T.; Wu, B.-R.; Liu, W.-C.; Hsieh, C.-Y.; Wu, P.-Y.; Sun, H.-Y.; Hung, C.-C. Immune reconstitution inflammatory syndrome of Kaposi’s sarcoma in an HIV-infected patient. J. Microbiol. Immunol. Infect. 2013, 46, 309–312. [Google Scholar] [CrossRef] [Green Version]
  70. Cheng, C.-H.; Hsu, Y.-H. Immune reconstitution inflammatory syndrome-associated disseminated Kaposi’s sarcoma in a patient infected with human immunodeficiency virus: Report of an autopsy case. Ci Ji Yi Xue Za Zhi 2017, 29, 41–45. [Google Scholar] [CrossRef] [Green Version]
  71. Lee, A.J.; Brenner, L.; Mourad, B.; Monteiro, C.; Vega, K.J.; Munoz, J.C. Gastrointestinal Kaposi’s sarcoma: Case report and review of the literature. World J. Gastrointest. Pharmacol. Ther. 2015, 6, 89–95. [Google Scholar] [CrossRef]
  72. Arora, M.; Goldberg, E.M. Kaposi sarcoma involving the gastrointestinal tract. Gastroenterol. Hepatol. 2010, 6, 459–462. [Google Scholar]
  73. Suresh, K.; Semaan, R.; Arias, S.; Karakousis, P.; Lee, H. Pleuropulmonary Kaposi Sarcoma in the Setting of Immune Reactivation. J. Pulm. Respir. Med. 2016, 6, 352. [Google Scholar] [CrossRef] [Green Version]
  74. Volkow, P.F.; Cornejo, P.; Zinser, J.W.; Ormsby, C.E.; Reyes-Terán, G. Life-threatening exacerbation of Kaposi’s sarcoma after prednisone treatment for immune reconstitution inflammatory syndrome. AIDS 2008, 22, 663–665. [Google Scholar] [CrossRef]
  75. Papagatsia, Z.; Jones, J.; Morgan, P.; Tappuni, A.R. Oral Kaposi sarcoma: A case of immune reconstitution inflammatory syndrome. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2009, 108, 70–75. [Google Scholar] [CrossRef]
  76. Silva, L.; Azurara, L.; Monteiro, A.F.; Miroux-Catarino, A.; Amaro, C.; Viana, I. Pediatric Kaposi’s sarcoma associated with immune reconstitution inflammatory syndrome. Pediatr. Dermatol. 2020, 37, 239–240. [Google Scholar] [CrossRef] [PubMed]
  77. Addula, D.; Das, C.J.; Kundra, V. Imaging of Kaposi sarcoma. Abdom. Radiol. 2021, 46, 5297–5306. [Google Scholar] [CrossRef] [PubMed]
  78. Dupin, N.; Jary, A.; Boussouar, S.; Syrykh, C.; Gandjbakhche, A.; Bergeret, S.; Palich, R. Current and Future Tools for Diagnosis of Kaposi’s Sarcoma. Cancers 2021, 13, 5927. [Google Scholar] [CrossRef] [PubMed]
  79. Crane, H.M.; Deubner, H.; Huang, J.C.; Swanson, P.E.; Harrington, R.D. Fatal Kaposi’s sarcoma-associated immune reconstitution following HAART initiation. Int. J. STD AIDS 2005, 16, 80–83. [Google Scholar] [CrossRef] [PubMed]
  80. Weir, A.; Wansbrough-Jones, M. Mucosal Kaposi’s sarcoma following protease inhibitor therapy in an HIV-infected patient. AIDS 1997, 11, 1895–1896. [Google Scholar] [CrossRef] [PubMed]
  81. Odongo, F.C.A. Fatal Disseminated Kaposi’s Sarcoma due to Immune Reconstitution Inflammatory Syndrome following HAART Initiation. Case Rep. Infect. Dis. 2013, 546578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Palmieri, C.; Dhillon, T.; Thirlwell, C.; Newsom-Davis, T.; Young, A.-M.; Nelson, M.; Gazzard, B.G.; Bower, M. Pulmonary Kaposi sarcoma in the era of highly active antiretroviral therapy. HIV Med. 2006, 7, 291–293. [Google Scholar] [CrossRef] [PubMed]
  83. Dumic, I.; Radovanovic, M.; Igandan, O.; Savic, I.; Nordstrom, C.W.; Jevtic, D.; Subramanian, A.; Ramanan, P. A Fatal Case of Kaposi Sarcoma Immune Reconstitution Syndrome (KS-IRIS) Complicated by Kaposi Sarcoma Inflammatory Cytokine Syndrome (KICS) or Multicentric Castleman Disease (MCD): A Case Report and Review. Am. J. Case Rep. 2020, 21, e926433. [Google Scholar] [CrossRef] [PubMed]
  84. Polizzotto, M.N.; Uldrick, T.S.; Wyvill, K.M.; Aleman, K.; Marshall, V.; Wang, V.; Whitby, D.; Pittaluga, S.; Jaffe, E.S.; Millo, C.; et al. Clinical Features and Outcomes of Patients With Symptomatic Kaposi Sarcoma Herpesvirus (KSHV)-associated Inflammation: Prospective Characterization of KSHV Inflammatory Cytokine Syndrome (KICS). Clin. Infect. Dis. 2016, 62, 730–738. [Google Scholar] [CrossRef]
  85. Karass, M.; Grossniklaus, E.; Seoud, T.; Jain, S.; Goldstein, D.A. Kaposi Sarcoma Inflammatory Cytokine Syndrome (KICS): A Rare but Potentially Treatable Condition. Oncologist 2017, 22, 623–625. [Google Scholar] [CrossRef] [Green Version]
  86. Goncalves, P.H.; Ziegelbauer, J.; Uldrick, T.S.; Yarchoan, R. Kaposi sarcoma herpesvirus-associated cancers and related diseases. Curr. Opin. HIV AIDS 2017, 12, 47–56. [Google Scholar] [CrossRef]
  87. Török, M.E.; Yen, N.T.B.; Chau, T.T.H.; Mai, N.T.H.; Phu, N.H.; Mai, P.P.; Dung, N.T.; Chau, N.V.V.; Bang, N.D.; Tien, N.A.; et al. Timing of initiation of antiretroviral therapy in human immunodeficiency virus (HIV)—associated tuberculous meningitis. Clin. Infect. Dis. 2011, 52, 1374–1383. [Google Scholar] [CrossRef]
  88. Dutertre, M.; Cuzin, L.; Demonchy, E.; Puglièse, P.; Joly, V.; Valantin, M.-A.; Cotte, L.; Huleux, T.; Delobel, P.; Martin-Blondel, G.; et al. Initiation of Antiretroviral Therapy Containing Integrase Inhibitors Increases the Risk of IRIS Requiring Hospitalization. J. Acquir. Immune Defic. Syndr. 2017, 76, e23–e26. [Google Scholar] [CrossRef]
  89. Kityo, C.; Szubert, A.J.; Siika, A.; Heyderman, R.; Bwakura-Dangarembizi, M.; Lugemwa, A.; Mwaringa, S.; Griffiths, A.; Nkanya, I.; Kabahenda, S.; et al. Raltegravir-intensified initial antiretroviral therapy in advanced HIV disease in Africa: A randomised controlled trial. PLoS Med. 2018, 15, e1002706. [Google Scholar] [CrossRef] [Green Version]
  90. Hill, A.M.; Mitchell, N.; Hughes, S.; Pozniak, A.L. Risks of cardiovascular or central nervous system adverse events and immune reconstitution inflammatory syndrome, for dolutegravir versus other antiretrovirals: Meta-analysis of randomized trials. Curr. Opin. HIV AIDS 2018, 13, 102–111. [Google Scholar] [CrossRef]
  91. Groupe d’Experts Sous la Direction du Pr Philippe Morlat Cancers. In Prise en Charge Medicale des Personnes Vivant Avec le VIH. August 2017. Available online: https://cns.sante.fr/actualites/prise-en-charge-du-vih-recommandations-du-groupe-dexperts/ (accessed on 8 February 2021).
  92. Yarchoan, R.; Uldrick, T.S. HIV-Associated Cancers and Related Diseases. N. Engl. J. Med. 2018, 378, 1029–1041. [Google Scholar] [CrossRef] [PubMed]
  93. Abbar, B.; Veyri, M.; Solas, C.; Poizot-Martin, I.; Spano, J.-P. HIV and cancer: Update 2020. Bull. Cancer 2020, 107, 21–29. [Google Scholar] [CrossRef] [PubMed]
  94. Gbabe, O.F.; Okwundu, C.I.; Dedicoat, M.; Freeman, E.E. Treatment of severe or progressive Kaposi’s sarcoma in HIV-infected adults. Cochrane Database Syst. Rev. 2014, CD003256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bower, M.; Weir, J.; Francis, N.; Newsom-Davis, T.; Powles, S.; Crook, T.; Boffito, M.; Gazzard, B.; Nelson, M. The effect of HAART in 254 consecutive patients with AIDS-related Kaposi’s sarcoma. AIDS 2009, 23, 1701–1706. [Google Scholar] [CrossRef] [PubMed]
  96. Mosam, A.; Shaik, F.; Uldrick, T.S.; Esterhuizen, T.; Friedland, G.H.; Scadden, D.T.; Aboobaker, J.; Coovadia, H.M. A randomized controlled trial of highly active antiretroviral therapy versus highly active antiretroviral therapy and chemotherapy in therapy-naive patients with HIV-associated Kaposi sarcoma in South Africa. J. Acquir. Immune Defic. Syndr. 2012, 60, 150–157. [Google Scholar] [CrossRef] [Green Version]
  97. Borok, M.; Fiorillo, S.; Gudza, I.; Putnam, B.; Ndemera, B.; White, I.E.; Gwanzura, L.; Schooley, R.T.; Campbell, T.B. Evaluation of plasma human herpesvirus 8 DNA as a marker of clinical outcomes during antiretroviral therapy for AIDS-related Kaposi sarcoma in Zimbabwe. Clin. Infect. Dis. 2010, 51, 342–349. [Google Scholar] [CrossRef] [Green Version]
  98. Krown, S.E.; Moser, C.B.; MacPhail, P.; Matining, R.M.; Godfrey, C.; Caruso, S.R.; Hosseinipour, M.C.; Samaneka, W.; Nyirenda, M.; Busakhala, N.W.; et al. Treatment of advanced AIDS-associated Kaposi sarcoma in resource-limited settings: A three-arm, open-label, randomised, non-inferiority trial. Lancet 2020, 395, 1195–1207. [Google Scholar] [CrossRef]
  99. Hosseinipour, M.C.; Kang, M.; Krown, S.E.; Bukuru, A.; Umbleja, T.; Martin, J.N.; Orem, J.; Godfrey, C.; Hoagland, B.; Mwelase, N.; et al. As-Needed Vs Immediate Etoposide Chemotherapy in Combination With Antiretroviral Therapy for Mild-to-Moderate AIDS-Associated Kaposi Sarcoma in Resource-Limited Settings: A5264/AMC-067 Randomized Clinical Trial. Clin. Infect. Dis. 2018, 67, 251–260. [Google Scholar] [CrossRef]
  100. Casper, C.; Krantz, E.M.; Corey, L.; Kuntz, S.R.; Wang, J.; Selke, S.; Hamilton, S.; Huang, M.-L.; Wald, A. Valganciclovir for suppression of human herpesvirus-8 replication: A randomized, double-blind, placebo-controlled, crossover trial. J. Infect. Dis. 2008, 198, 23–30. [Google Scholar] [CrossRef]
  101. Martin, D.F.; Kuppermann, B.D.; Wolitz, R.A.; Palestine, A.G.; Li, H.; Robinson, C.A. Oral ganciclovir for patients with cytomegalovirus retinitis treated with a ganciclovir implant. Roche Ganciclovir Study Group. N. Engl. J. Med. 1999, 340, 1063–1070. [Google Scholar] [CrossRef]
  102. Jones, J.L.; Hanson, D.L.; Chu, S.Y.; Ward, J.W.; Jaffe, H.W. AIDS-associated Kaposi’s sarcoma. Science 1995, 267, 1078–1079. [Google Scholar] [CrossRef] [PubMed]
  103. Mocroft, A.; Youle, M.; Gazzard, B.; Morcinek, J.; Halai, R.; Phillips, A.N. Anti-herpesvirus treatment and risk of Kaposi’s sarcoma in HIV infection. Royal Free/Chelsea and Westminster Hospitals Collaborative Group. AIDS 1996, 10, 1101–1105. [Google Scholar] [PubMed]
  104. Glesby, M.J.; Hoover, D.R.; Weng, S.; Graham, N.M.; Phair, J.P.; Detels, R.; Ho, M.; Saah, A.J. Use of antiherpes drugs and the risk of Kaposi’s sarcoma: Data from the Multicenter AIDS Cohort Study. J. Infect. Dis. 1996, 173, 1477–1480. [Google Scholar] [CrossRef] [Green Version]
  105. Spector, S.A.; McKinley, G.F.; Lalezari, J.P.; Samo, T.; Andruczk, R.; Follansbee, S.; Sparti, P.D.; Havlir, D.V.; Simpson, G.; Buhles, W.; et al. Oral ganciclovir for the prevention of cytomegalovirus disease in persons with AIDS. Roche Cooperative Oral Ganciclovir Study Group. N. Engl. J. Med. 1996, 334, 1491–1497. [Google Scholar] [CrossRef] [PubMed]
  106. Volkow, P.; Chávez-Galán, L.; Ramón-Luing, L.; Cruz-Velazquez, J.; Cornejo-Juárez, P.; Sada-Ovalle, I.; Pérez-Padilla, R.; Islas-Muñoz, B. Impact of valganciclovir therapy on severe IRIS-Kaposi sarcoma attributable mortality. MedRxiv 2020. [Google Scholar] [CrossRef]
  107. Krown, S.E.; Metroka, C.; Wernz, J.C. Kaposi’s sarcoma in the acquired immune deficiency syndrome: A proposal for uniform evaluation, response, and staging criteria. AIDS Clinical Trials Group Oncology Committee. J. Clin. Oncol. 1989, 7, 1201–1207. [Google Scholar] [CrossRef]
  108. Krown, S.E.; Testa, M.A.; Huang, J. AIDS-related Kaposi’s sarcoma: Prospective validation of the AIDS Clinical Trials Group staging classification. AIDS Clinical Trials Group Oncology Committee. J. Clin. Oncol. 1997, 15, 3085–3092. [Google Scholar] [CrossRef]
  109. Guo, W.X.; Antakly, T. AIDS-related Kaposi’s sarcoma: Evidence for direct stimulatory effect of glucocorticoid on cell proliferation. Am. J. Pathol. 1995, 146, 727–734. [Google Scholar]
  110. Schulhafer, E.P.; Grossman, M.E.; Fagin, G.; Bell, K.E. Steroid-induced Kaposi’s sarcoma in a patient with pre-AIDS. Am. J. Med. 1987, 82, 313–317. [Google Scholar] [CrossRef]
  111. Pourcher, V.; Desnoyer, A.; Assoumou, L.; Lebbe, C.; Curjol, A.; Marcelin, A.-G.; Cardon, F.; Gibowski, S.; Salmon, D.; Chennebault, J.-M.; et al. Phase II Trial of Lenalidomide in HIV-Infected Patients with Previously Treated Kaposi’s Sarcoma: Results of the ANRS 154 Lenakap Trial. AIDS Res. Hum. Retrovir. 2017, 33, 1–10. [Google Scholar] [CrossRef]
  112. Uldrick, T.S.; Wyvill, K.M.; Kumar, P.; O’Mahony, D.; Bernstein, W.; Aleman, K.; Polizzotto, M.N.; Steinberg, S.M.; Pittaluga, S.; Marshall, V.; et al. Phase II study of bevacizumab in patients with HIV-associated Kaposi’s sarcoma receiving antiretroviral therapy. J. Clin. Oncol. 2012, 30, 1476–1483. [Google Scholar] [CrossRef] [PubMed]
  113. Masood, R.; Cai, J.; Zheng, T.; Smith, D.L.; Naidu, Y.; Gill, P.S. Vascular endothelial growth factor/vascular permeability factor is an autocrine growth factor for AIDS-Kaposi sarcoma. Proc. Natl. Acad. Sci. USA 1997, 94, 979–984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Dezube, B.J.; Krown, S.E.; Lee, J.Y.; Bauer, K.S.; Aboulafia, D.M. Randomized phase II trial of matrix metalloproteinase inhibitor COL-3 in AIDS-related Kaposi’s sarcoma: An AIDS Malignancy Consortium Study. J. Clin. Oncol. 2006, 24, 1389–1394. [Google Scholar] [CrossRef]
  115. Koon, H.B.; Bubley, G.J.; Pantanowitz, L.; Masiello, D.; Smith, B.; Crosby, K.; Proper, J.; Weeden, W.; Miller, T.E.; Chatis, P.; et al. Imatinib-induced regression of AIDS-related Kaposi’s sarcoma. J. Clin. Oncol. 2005, 23, 982–989. [Google Scholar] [CrossRef] [PubMed]
  116. Vart, R.J.; Nikitenko, L.L.; Lagos, D.; Trotter, M.W.B.; Cannon, M.; Bourboulia, D.; Gratrix, F.; Takeuchi, Y.; Boshoff, C. Kaposi’s sarcoma-associated herpesvirus-encoded interleukin-6 and G-protein-coupled receptor regulate angiopoietin-2 expression in lymphatic endothelial cells. Cancer Res. 2007, 67, 4042–4051. [Google Scholar] [CrossRef] [Green Version]
  117. Krown, S.E.; Roy, D.; Lee, J.Y.; Dezube, B.J.; Reid, E.G.; Venkataramanan, R.; Han, K.; Cesarman, E.; Dittmer, D.P. Rapamycin with antiretroviral therapy in AIDS-associated Kaposi sarcoma: An AIDS Malignancy Consortium study. J. Acquir. Immune Defic. Syndr. 2012, 59, 447–454. [Google Scholar] [CrossRef] [Green Version]
  118. Galanina, N.; Goodman, A.M.; Cohen, P.R.; Frampton, G.M.; Kurzrock, R. Successful Treatment of HIV-Associated Kaposi Sarcoma with Immune Checkpoint Blockade. Cancer Immunol. Res. 2018, 6, 1129–1135. [Google Scholar] [CrossRef] [Green Version]
  119. Dalla Pria, A.; Pinato, D.J.; Bracchi, M.; Bower, M. Recent advances in HIV-associated Kaposi sarcoma. F1000Res 2019, 8, 970. [Google Scholar] [CrossRef] [Green Version]
  120. Cesmeci, E.; Guven, D.C.; Aktas, B.Y.; Aksoy, S. Case of metastatic kaposi sarcoma successfully treated with anti-PD-1 immunotherapy. J. Oncol. Pharm. Pract. 2021, 27, 1766–1769. [Google Scholar] [CrossRef]
  121. Barrios, D.M.; Do, M.H.; Phillips, G.S.; Postow, M.A.; Akaike, T.; Nghiem, P.; Lacouture, M.E. Immune checkpoint inhibitors to treat cutaneous malignancies. J. Am. Acad. Dermatol. 2020, 83, 1239–1253. [Google Scholar] [CrossRef]
  122. Cook, M.R.; Kim, C. Safety and Efficacy of Immune Checkpoint Inhibitor Therapy in Patients With HIV Infection and Advanced-Stage Cancer: A Systematic Review. JAMA Oncol. 2019, 5, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  123. Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O.; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. Immunother. Cancer 2017, 5, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Pantanowitz, L.; Früh, K.; Marconi, S.; Moses, A.V.; Dezube, B.J. Pathology of rituximab-induced Kaposi sarcoma flare. BMC Clin. Pathol. 2008, 8, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Uldrick, T.S.; Gonçalves, P.H.; Abdul-Hay, M.; Claeys, A.J.; Emu, B.; Ernstoff, M.S.; Fling, S.P.; Fong, L.; Kaiser, J.C.; Lacroix, A.M.; et al. Assessment of the Safety of Pembrolizumab in Patients With HIV and Advanced Cancer-A Phase 1 Study. JAMA Oncol. 2019, 5, 1332–1339. [Google Scholar] [CrossRef]
Table 1. General IRIS case definitions.
Table 1. General IRIS case definitions.
According to French et al., 2004 [24]
Diagnosis Requires Two Major Criteria (A+B) or Major Criterion (A) Plus Two Minor Criteria to Be Fulfilled
Major criteriaMinor criteria
(A)
Atypical presentation of opportunistic infections or tumors in patients responding to ART
  • Increase in blood CD4 T-cell count after starting ART
-
Localized disease
-
Exaggerated inflammatory reaction
-
Atypical inflammatory response in affected tissues
-
Progressive organ dysfunction or enlargement of pre-existing lesions after definite clinical improvement with pathogen-specific therapy before the initiation of ART and exclusion of treatment toxicity and new alternative diagnoses
  • Increase in an immune response specific to the relevant pathogen—e.g., delayed type hypersensitivity skin test response to mycobacterial antigens
(B)
Decrease in plasma HIV RNA concentration by more than 1 log10 copies per mL
  • Spontaneous resolution of disease without specific antimicrobial therapy or tumor chemotherapy with continuation of ART
According to Shelburne et al., 2006 [31]
Criteria for IRIS Diagnosis Include:
  • HIV-infected patient
  • Receiving effective ART as evidenced by a decrease in HIV-[1] RNA concentration from baseline or an increase in CD4+ T cells from baseline (may lag behind HIV-1 RNA decrease)
  • Clinical symptoms consistent with inflammatory process
  • Clinical course not consistent with expected course of previously diagnosed opportunistic infection, expected course of newly diagnosed opportunistic infection, or drug toxicity
Table 2. Staging classification for AIDS-related KS (Krown et al., 1989) [107].
Table 2. Staging classification for AIDS-related KS (Krown et al., 1989) [107].
CriteriaGood Risk (All of the Following)Poor Risk (Any of the Following)
Tumor, TT0: confined to the skin and/or lymph nodes and/or minimal oral disease (non-nodular KS confined to palate)T1: Tumor-associated edema or ulceration
Extensive oral KS
Gastrointestinal KS
KS in organs other than lymph nodes
Immune system, I 1I0: CD4 T-cell count ≥150/mm3I1: CD4T cell <150/mm3
Systemic disease, SS0: No history of opportunistic infection or thrush
No “B symptoms” 2
Karnofsky performance status ≥70
S1: History of opportunistic infection and/or thrush
“B” symptoms present
Karnofsky performance status <70
Other HIV-related illness (eg, neurologic disease, lymphoma)
1 I stage has less prognostic value than T or S stages in the presence of ART therapy 2 “B” symptoms are unexplained fever, night sweats, >10 percent involuntary weight loss, or diarrhea persisting more than two weeks.
Table 3. Disease progression criteria according to ACTG response criteria (Krown et al., 1989) [107].
Table 3. Disease progression criteria according to ACTG response criteria (Krown et al., 1989) [107].
-
An increase of 25% or more in the size of previously existing lesions
-
The appearance of new lesions or new sites of disease
-
A change in the character of 25% or more of the skin or oral lesions from macular to plaque-like or nodular
-
The development of new or increasing tumor- associated edema or effusions
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Poizot-Martin, I.; Brégigeon, S.; Palich, R.; Marcelin, A.-G.; Valantin, M.-A.; Solas, C.; Veyri, M.; Spano, J.-P.; Makinson, A. Immune Reconstitution Inflammatory Syndrome Associated Kaposi Sarcoma. Cancers 2022, 14, 986. https://doi.org/10.3390/cancers14040986

AMA Style

Poizot-Martin I, Brégigeon S, Palich R, Marcelin A-G, Valantin M-A, Solas C, Veyri M, Spano J-P, Makinson A. Immune Reconstitution Inflammatory Syndrome Associated Kaposi Sarcoma. Cancers. 2022; 14(4):986. https://doi.org/10.3390/cancers14040986

Chicago/Turabian Style

Poizot-Martin, Isabelle, Sylvie Brégigeon, Romain Palich, Anne-Geneviève Marcelin, Marc-Antoine Valantin, Caroline Solas, Marianne Veyri, Jean-Philippe Spano, and Alain Makinson. 2022. "Immune Reconstitution Inflammatory Syndrome Associated Kaposi Sarcoma" Cancers 14, no. 4: 986. https://doi.org/10.3390/cancers14040986

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