Abstract
The advent of molecularly targeted agents for patients with peripheral T-cell lymphomas (PTCL) has begun to change the therapeutic landscape in these diseases, especially for patients with relapsed or refractory disease. These agents, grounded in targeting numerous pathways or alterations related to disease pathogenesis, have shown promise across many PTCL sub-histologies. Aided by significant advances in experimental techniques related to molecular biology, epigenetics, and immunology, more recent studies have begun elucidating mediators of resistance, both intrinsic and acquired, to inform future therapeutic advances. Defining and targeting these escape mechanisms through rational combination approaches will likely be important to continue to build on these promising advances and further improve clinical outcomes for patients facing PTCL.
Keywords: Targeted therapies, Kinase Inhibitors, Epigenetic Therapies, Immunomodulatory Therapies, Peripheral T-cell lymphomas
Introduction
Peripheral T-cell lymphomas (PTCL) comprise a heterogeneous collection of malignancies arising from the malignant transformation of mature T cells and include nearly 30 histologic subtypes with distinct clinical behavior and disease pathogenesis. Patients with aggressive PTCL subtypes are frequently treated with upfront combination chemotherapy with consideration of stem cell transplantation if remission is achieved depending on the specific disease subtype and patient suitability [1]. Other patients ineligible for this intensive approach or with more indolent disease behavior are typically treated with ongoing, sequential, maintenance-type treatments attempting to maximize clinical benefit with acceptable long-term toxicities. Broadly, however, compared to those for patients with B-cell lymphomas, the outcomes for patients with PTCL are poor, especially for those patients with relapsed or refractory (R/R) disease [2, 3]. This disparity has stimulated significant recent progress in defining the disease pathobiology of PTCL subtypes and the development of novel therapeutic approaches to ultimately improve patient outcomes.
The advent of high-throughput genomic sequencing and advances in molecular biology have generated deep insights into the genomic landscape and molecular pathogenesis of PTCL subtypes. Indeed, nearly all PTCL sub-histologies have multiple published manuscripts detailing the landscape of molecular alterations present [4–8]. For example, Watatani and colleagues used whole-exome sequencing on PTCL tumor samples to reveal a new molecular subtype of PTCL-NOS characterized by alterations in TP53 and CDKN2A. Paralleling these discoveries into the genetic lesions underlying PTCL has been other lines of investigation into the cellular signaling pathways that PTCLs are dependent on for growth and proliferation [9, 10] that may be susceptible to therapeutic targeting. Among these that are most advanced therapeutically are the phosphatidyl inositol-3-kinase pathway (PI3K), the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, and the spleen tyrosine kinase (SYK) pathway.
An additional line of background research in PTCL pathogenesis concerns epigenetic alterations in PTCL. Many PTCL subtypes contain epigenetic changes and/or mutations in epigenetic-related genes, which has prompted the use of epigenetic-directed therapies such as histone deacetylase (HDAC) inhibitors [11]. Besides implications for pathogenesis, such markers may carry prognostic importance [12, 13]. Epigenetic alterations are especially well defined in the PTCL-TFH/AITL subtypes with frequent mutations in epigenetic regulators such as TET2 and DNMT3A [7], and emerging evidence suggests superior efficacy with HDAC inhibition in these diseases compared to other PTCL subtypes [14]. Newer targeted approaches are seeking to explore other epigenetic-targeted therapies such as the enhancer of zeste homolog 1/2 (EZH1/2) [15] or non-enzyme inhibition-based approaches [16, 17] to disrupting perturbed epigenetic networks and pathways for therapeutic gain.
In the present review, we discuss therapeutic targets and compounds under investigation for use in treating R/R PTCL-NOS, focusing on those farthest along in clinical development or showing particular promise in earlier-phase investigations. Where possible, we will seek to highlight translational efforts seeking to understand mediators of response to these agents.
Kinase-targeted therapies
PI3K
Cellular signaling through the PI3K-Akt-molecular target of rapamycin (mTOR) pathway has been shown to be critical for growth and differentiation of lymphocytes [9, 18, 19], and therefore targeting this pathway with small molecule PI3K inhibitors has emerged as a promising approach in many malignancies, especially lymphoid cancers. Available PI3K inhibitors include idelalisib, copanlisib, zandelisib, tenalisib, and duvelisib; the last two agents are most investigated for patients with R/R PTCL [20, 21]. A completed phase I study of duvelisib, a, oral δ/γ isoform-specific inhibitor, showed promising responses among 16 patients with R/R PTCL (study also included patients with R/R cutaneous T-cell lymphomas, not discussed here) following a median of >2 lines of therapy. In this population (largest histologic subtype PTCL-NOS, N = 6), the overall response rate was 50%, including 3 complete responses and 5 partial responses. Analysis of toxicities observed among all patients showed side effects common in this class of agents: elevations in AST/ALT (31% grade >2), rash (14% grade >2), and grades 1/2 pyrexia (37%) and cough (34%). Biologically, increases in CCL1, IL-17a, and sCD40L and diminution in p40 and CXCL13 correlated with responses. This has prompted the ongoing phase II dose optimization/expansion study (NCT03372057) seeking to further explore and potentially register the use of this agent in patients with R/R PTCL [22]. In preliminary results among 25 response assessed patients, 13 had responses, including 9 complete responses, with a median duration of response of 4.1 months. The toxicity profile was notable for cytopenias (24%) and elevated ALT (56%).
More recently, phase I data concerning another dual PI3K δ/γ inhibitor tenalisib were published including 28 patients with R/R PTCL (histologic subtypes not reported), over 20% of whom had received >4 prior lines of therapy [21]. Among these 28 patients, 15 completed at least 2 cycles of therapy, 3 of whom achieved CR and 4 achieved PR. The median duration of response was 6.5 months. Toxicities were generally comparable to those observed with duvelisib, with the most common grade >2 toxicity being elevated AST/ALT (19% each across all patients). Based on these promising results, two ongoing studies are investigating each of these agents in combination with the HDAC inhibitor romidepsin (NCT02783625 and NCT03770000) for patients with R/R PTCL. Other ongoing or planned studies with PI3K inhibitors in patients with R/R PTCL includes combination with immune checkpoint blockade (copanlisib plus pembrolizumab, NCT02535247) and with chemotherapy (copanlisib plus gemcitabine, NCT03052933). For upfront treatment, a planned intergroup study (A051902) will randomize patients with CD30-negative PTCL to CHO(E)P versus CHO(E)P plus duvelisib versus CHO(E)P plus azacitidine to advance these promising agents earlier in the care of patients.
JAK
The Janus kinase family proteins (JAK1, JAK2, JAK3, and TYK2) contribute to critical pathways mediating immune responses and inflammation in normal physiology. Downstream of the JAK proteins are the STAT family of transcription factors, most importantly STAT3 and STAT5B, which mediate effects of JAK signaling through transcriptional regulation. Cancer-associated JAK family gene mutations were initially discovered in the myeloproliferative neoplasms, but recent work has begun elucidating their prevalence and significance across nearly all TCL histologies and potential for therapeutic targeting [23–28]. Besides activating mutations, there are also gene fusions involving JAK family genes [29] leading to activated JAK-STAT pathway signaling. Recent data have emerged regarding efficacy of JAK pathway inhibition as a therapeutic approach in PTCL: Moskowitz and colleagues reported results from a phase II study of ruxolitinib in 53 patients with R/R PTCL, with 48 evaluable patients. The largest subgroup by histology was PTCL-NOS (N = 11) followed by PTCL-TFH and angioimmunoblastic T-cell lymphoma (AITL, N = 9). Patients were analyzed according to JAK/STAT pathway status: activating mutation present, pathway activation evident by immunohistochemistry, or neither. The overall response rate (including patients with cutaneous T-cell lymphomas) was 23%; the response rate plus rate of maintaining stable disease for >6 months reached 35%. Three patients achieved complete responses, and 8 (17%) partial responses. In terms of subtypes, particularly high rates of clinical benefit were seen in patients with PTCL-TFH/AITL (44%), large granular lymphocytic leukemia (75%), and T-cell prolymphocytic leukemia (50%). When analyzed according to JAK/STAT pathway status, the overall response rate was 29% in those with an activating mutation present or elevated pSTAT3 by immunohistochemistry compared to only 11% in those with neither. The most frequent drug-related grade 3/4 adverse events were cytopenias, with neutropenia being the most common (N = 13). An ongoing study (NCT01712659) is testing ruxolitinib specifically in patients with adult T-cell leukemia/lymphoma harboring JAK/STAT pathway activation [28, 30].
SYK
SYK encodes a protein tyrosine kinase with pleiotropic downstream signaling partners, including PI3K and PLC-gamma [31], and is normally expressed on B cells but absent on T cells [32]. Aberrant expression of SYK is observed in the majority (94%) of T-cell lymphoma (TCL) cases [33]. Cerdulatinib is a pan-JAK/SYK pathway inhibitor that has been investigated for treating patients with R/R hematologic malignancies, including PTCL [34]. Among 60 evaluable patients with PTCL who received cerdulatinib, the overall response rate was 35%; the response rate specifically in patients with PTCL-TFH/AITL was 55% (12/22). The main treatment-emergent adverse event grade >2 included lipase elevation (21%) with diarrhea, neutropenia, anemia, and fatigue all between 5–10%. Another SYK inhibitor, TAK-659, which also targets fms-like tyrosine kinase 3, is in development and is undergoing evaluation for patients with B-cell non-Hodgkin lymphomas (NCT03357627, NCT02000934, and NCT03742258).
ITK
The interleukin-2-inducible kinase (ITK) is a member of the Tec family non-receptor tyrosine kinase and mediates T-cell activation and differentiation [35]. ITK is specific to T cells and is required for signaling through the T-cell receptor: when TCR is activated, ITK is recruited, phosphorylated, and activated, thus activating secondary downstream messengers including members of the NF-κB, mTOR, and ERK pathways [36]. Certain PTCL subtypes have been shown to harbor activating ITK fusions [37] and pre-clinical evidence suggests ITK-mediating signaling may underlie chemoresistance [38]. This has spurred the development of small molecule inhibitors of ITK for use in PTCL, including CPI-818. Interim results for this agent [39] included 16 patients in dose escalation with a variety of PTCL and cutaneous T-cell lymphoma sub-histologies (including 4 patients with PTCL-NOS). Evidence of activity with no grade 3/4 adverse events has been noted particularly in patients with Sezary Syndrome, a form of cutaneous T-cell lymphoma, and the study remains ongoing (NCT03952078).
Aurora A kinase
Aurora kinase A is a serine/threonine protein kinase that mediates spindle formation during mitosis and is overexpressed in a wide range of malignancies, including PTCL [40], and targeting this pathway was shown to have pre-clinical activity [41]. Alisertib is an oral Aurora kinase A inhibitor, and data for this agent came from a randomized, phase III study of alisertib versus investigator’s choice in R/R PTCL [42]. The study was terminated early due to evidence of lack of benefit for progression-free survival for alisertib, with an overall response rate of 33% compared to 45% for the comparison arm. Based on these results, further single-agent development has been halted for patients with PTCL.
ALK
Anaplastic large cell lymphoma (ALCL) is a common subtype of PTCL and is subdivided based on the presence or absence of the t(2;5)(p23;q35) translocation that fuses the anaplastic lymphoma kinase (ALK) gene to NPM. As ALK is a readily targetable tyrosine kinase receptor, this has led to use of ALK inhibitors for patients with R/R ALK+ ALCL [43, 44]. Crizotinib, the first ALK inhibitor developed, was used in 9 patients with R/R ALK+ ALCL, and all 9 patients experienced complete responses to therapy, many of which were durable [43]. Three of these 9 patients had prior stem cell transplantation (2 autologous, 1 allogeneic). Richly, et al. investigated the second-generation ALK-inhibitor ceritinib in 3 patients with R/R ALK+ ALCL, 2 of whom achieved CR, 1 PR, all of which were ongoing at time of publication [44].
Epigenetic-targeted therapies:
HDAC
Protein acetylation is governed by histone acetyltransferases that catalyze the addition of acetyl groups and HDAC enzymes, which remove them. Among HDAC inhibitors, belinostat [45] and romidepsin [46] are the most thoroughly studied for patients with PTCL histologies and HDAC inhibition is an established treatment for patients with R/R PTCL in current practice. Coiffier and colleagues reported updated results for romidepsin in patients with R/R PTCL [46] in 2014, involving 130 patients with PTCL-NOS, AITL, and ALK-negative anaplastic large cell lymphoma. The overall response rate was 25%, including 29% in the 69 patients with PTCL-NOS. The median time to response was 1.8 months and median duration of response was 28 months, highlighting its potential role as a chronic, maintenance-type therapy in this setting. The most common side effects included gastrointestinal toxicities (nausea, vomiting), cytopenias (especially anemia, thrombocytopenia), and fatigue/asthenia. Belinostat, among 129 patients with R/R PTCL, garnered a response rate of 26%; median progression-free survival and overall survival were 1.6 and 7.9 months, respectively. A similar toxicity profile to romidepsin was observed with this agent. Reported combination studies using romidepsin in R/R PTCL are summarized in Table 1.
Table 1:
Duvelisib | 14 | 36% | 21% | [47] |
Pembrolizumab | 15 | 44% | 20% | [48] |
Pralatrexate | 14 | 71% | 29% | [49] |
Liposomal doxorubicin | 12 | 25% | 25% | [50] |
Gemcitabine | 20 | 30% | 15% | [51] |
ICE | 18 | 93% | 80% | [52] |
Azacitidine | 6 | 83% | 50% | [53] |
Gemcitabine, cisplatin | 20 | 50% | 0% | [54] |
Lenalidomide, carfilzomib | 11 | 46% | 36% | [55] |
ICE, ifosfamide, carboplatin, and etoposide
Further studies are underway or planned to investigate other therapies in combination with HDAC inhibitors, including PI3K inhibitors (duvelisib, NCT02783625; tenalisib, NCT03770000), proteasome inhibitors (ixazomib, NCT03547700), aurora kinase inhibitors (alisertib, NCT01897012 [56]), other epigenetic therapies (azacitidine + lenalidomide NCT04447027), immunotherapy (durvalumab + pralatrexate or azacitidine, NCT03161223).
EZH1/2
The EZH1/2 enzymes are histone methyltransferases and serve as the active SET domain-containing catalytic subunits of the polycomb repressive complex 2, which regulates chromatin topology (reviewed in [57]). Mutations in EZH2 were observed in 10% (7/68) patients with hepatosplenic T-cell lymphoma [4], a rare and aggressive subtype of PTCL, and pre-clinical data suggest efficacy for EZH2 inhibition in this disease [58]. In follicular lymphoma, an indolent subtype of B-cell non-Hodgkin lymphoma, the EZH2 inhibitor tazemetostat has received accelerated FDA approval for patients with R/R disease, irrespective of EZH2 mutation status [59]. Fewer pre-clinical data exist concerning the role for EZH1/2 in PTCL. Dhiran and colleagues showed EZH2 IHC staining to be a distinguishing factor between malignant and normal T cells [60] and it was also shown that EZH2 overexpression is present in many TCL subtypes [61], mediated by the TCR/CD28 pathway. Emerging data suggest efficacy of EZH1/2 inhibition in R/R PTCL subtypes [15] with the dual EZH1/2 inhibitor valemetostat: among 9 patients with ATLL, there was 1 unconfirmed complete remission, 3 partial responses, and 3 patients with stable disease. Frequent adverse events included cytopenias (most commonly thrombocytopenia, 78%), dysgeusia, alopecia, and dry skin. An ongoing study (NCT02732275) will report further data for this promising agent more broadly in patients with PTCL.
DNMT
DNA methylation is a critical process to regulating gene expression and governed by tightly regulated enzymes that add or remove methyl groups at particular sequences. The DNA methyltransferase (DNMT) enzymes specifically catalyze the addition of methyl groups, and oncogenic lesions in these enzymes are principally found in PTCL-TFH and AITL subtypes within PTCL [6, 7]. This has spurred investigations into the use of DNMT inhibitors, also known as hypomethylating agents, for therapeutic gain in these diseases [53, 62–65], although most such reports are confined to single case reports or small case series. Falchi and colleagues reported data from a prospective study of 14 patients with R/R PTCL (65% with AITL) using the DNMT inhibitor 5-azacitidine plus romidepsin [53]. Among 13 response evaluable patients (completing ≥2 cycles of treatment), 7 (54%) experienced response, including 5 complete response and 2 partial responses. Among all patients, grade ≥3 treatment-emergent toxicities were primarily hematologic in nature. An ongoing upfront study [64] (NCT03542266) is evaluating oral azacitidine with combination chemotherapy for patients with PTCL, enriching for those with PTCL-TFH/AITL subtypes. Finally, an ongoing randomized study (NCT03593018) is evaluating oral azacitidine versus investigator’s choice in patients with R/R AITL.
Non-cell signaling kinase/non-epigenetic targets
Anti-apoptotic therapies
The advent of apoptosis-targeting therapies for patients with non-Hodgkin lymphoma has shown particular promise for patients with chronic lymphocytic leukemia and mantle cell lymphoma, with high rates of remission seen in each histology with the BCL2 inhibitor venetoclax [66, 67].
Emerging evidence suggests a role for such agents for treating patients with PTCL [68–70]. Apoptosis is governed by the BCL2 family of proteins with counterbalancing pro- and anti-apoptotic members, including MCL1 and BCL2. Spinner and colleagues revealed high levels of MCL1 expression across PTCL subtypes and that targeting this pathway delayed PTCL development and reduced survival in vivo [68]. Building off this work, Koch and colleagues identified an MCL1-dependent PTCL patient-derived tumor xenograft model wherein MCL1 inhibition improved survival and was synergistic with standard multiagent chemotherapy [70]. These data have spurred development of agents for patients with R/R PTCL in ongoing trials targeting MCL1 (PRT1419 – NCT04543305; AMG 397 – NCT03465540; AZD5991 – NCT03218683) and BCL2 (venetoclax – NCT03534180).
Stapled peptide therapies
Stapled peptides represent a novel class of anti-cancer agents that disrupt protein-protein interactions for therapeutic benefit, in contrast to enzymatic inhibition characteristic of most targeted agents. Preliminary data have been reported [71] in 26 patients with R/R PTCL by Shustov and colleagues using ALRN-6924. This agent mimics the inhibitor binding region of p53, thus binding endogenous inhibitors of p53 and restoring its normal induction of cell cycle arrest and apoptosis. In 15 evaluable patients, the overall response rate was 27% and disease control rate (ORR + SD) 47%, with an acceptable profile (most common treatment-related adverse event fatigue, 50% followed by nausea, 43%). Further development of this agent for patients with R/R PTCL are not currently planned.
Immunomodulatory agents
The immunomodulatory agent lenalidomide has broad activity across numerous hematologic malignancies, including multiple myeloma, deletion 5q myelodysplastic syndrome, mantle cell lymphoma, and follicular lymphoma. Lenalidomide binds to cereblon, the substrate adapter of the CRL4-cereblon E3 ubiquitin ligase complex, thus modulating its substrate specificity and inducing degradation of selected protein targets. As many of these protein targets mediate immune responses such as interleukin production and T/NK-cell proliferation/activation, these effects are believed to mediate the anti-tumor properties of this agent (reviewed in [72]). Furthermore, lenalidomide has been shown to strengthen immune synapses as another means to promote anti-tumor immunity [73]. Lenalidomide has been investigated for use in patients with R/R PTCL [74, 75] and is the subject of ongoing trials. Morschhauser and colleagues reported [74] an overall response rate of 22% in 54 patients with PTCL (median prior therapies 3); the median progression-free survival was 2.5 months. Frequent adverse events included thrombocytopenia (20%) and neutropenia (15%). Further data [75] were reported in 40 patients with a range of PTCL histologies, including 14 with PTCL-NOS. The overall response rate was similar, 26%. Ongoing investigations of lenalidomide in patients with R/R PTCL include in combination with the antibody-drug conjugate targeting CD30 brentuximab vedotin (NCT03302728, also including patients with Hodgkin lymphoma) and the anti-PD1 monoclonal antibody durvalumab (NCT03011814, also including patients with cutaneous T-cell lymphomas).
Farnesyltransferase inhibitors
Tipifarnib is a farnesyltransferase/CXCR4 inhibitor that has recently been investigated specifically for biomarker-driven use in patients with R/R PTCL, especially AITL or CXCL12+ PTCL-NOS [76]. CXCL12 expression has been suggested to be prognostic in patients with PTCL and tipifarnib was shown to downregulate CXCL12 secretion in stromal cultures, hence its selected use for treating patients with CXCL12+ PTCL [77, 78]. Within this trial, in the PTCL-NOS subcategory, patients were stratified according to the variant status of the 3’ UTR of the CXCL12 gene; among 9 evaluable patients with wildtype 3’ UTR, the clinical benefit rate was 82%, whereas all 6 patients with variant 3’ UTR had progressive disease [76]. The primary toxicities were cytopenias, especially thrombocytopenia (39%) and neutropenia (31%).
Incorporating molecularly targeted therapies into clinical practice
The care of patients with R/R PTCL is complex and a full discussion of this clinical scenario is beyond the scope of this review. However, in our practice, our approach relies principally on the patient’s goals/wishes for treatment and age/comorbidities, prior therapies received and tolerability therein, the tempo and extent of disease, and in some instances, availability of a suitable donor for allogeneic stem cell transplantation. Whenever feasible, we seek to molecularly characterize a patient’s tumor (preferably using a specimen confirming R/R disease) through targeted next-generation sequencing. Emerging evidence suggests that such results may carry implications for prognosis [79] or therapeutic selection [80]. In practice, we decide between non-cross-resistant combination therapies and targeted agents (potentially in the context of a clinical trial) primarily based on the tempo/extent of disease and the patient’s preferences for treatment, balancing what may be more quickly achieving a response versus a greater potential for lasting responses with chronic, maintenance-type treatments. Ongoing clinical trials mentioned in this article are summarized in Tables 2 and 3 (current as of January, 2021).
Table 2:
Kinase | Duvelisib | NCT03372057 | Recruiting | PTCL-NOS, AITL, ALCL, NKTL |
Duvelisib + romidepsin | NCT02783625 | Recruiting | PTCL, CTCL | |
Tenalisib | NCT03770000 | Active, not recruiting | TCL | |
Copanlisib + pembrolizumab | NCT02535247 | Active, not recruiting | PTCL, transformed CTCL | |
Copanlisib + gemcitabine | NCT03052933 | Active, not recruiting | PTCL, NKTL | |
Ruxolitinib | NCT01712659 | Recruiting | ATL | |
TAK-659 + venetoclax | NCT03357627 | Active, not recruiting | NHL | |
TAK-659 | NCT02000934 | Active, not recruiting | Lymphoma | |
CPI-818 | NCT03952078 | Recruiting | TCL | |
Tenalisib + romidepsin | NCT03770000 | Active, not recruiting | TCL | |
Epigenetic | Romidepsin + ixazomib | NCT03547700 | Active, not recruiting | PTCL |
Romidepsin + azacitidine + lenalidomide | NCT04447027 | Recruiting | TCL | |
Romidepsin +/− pralatrexate +/− durvalumab +/− azacitidine | NCT03161223 | Recruiting | PTCL | |
Valemetostat | NCT02732275 | Recruiting | NHL | |
Azacitidine | NCT03593018 | Recruiting | AITL | |
Other | PRT1419 | NCT04543305 | Recruiting | NHL |
AMG 397 | NCT03465540 | Recruiting | NHL | |
AZD5991 | NCT03218683 | Recruiting | NHL | |
Venetoclax | NCT03534180 | Recruiting | PTCL, transformed MF | |
Lenalidomide + brentuximab | NCT03302728 | Recruiting | CD30+ PTCL | |
Lenalidomide + durvalumab | NCT03011814 | Recruiting | PTCL |
NKTL, NK/T-cell lymphoma; CTCL, cutaneous T-cell lymphoma; FTCL, follicular T-cell lymphoma; ATL, adult T-cell leukemia/lymphoma
Table 3:
Kinase | Cerdulatinib | Phase I ongoing |
TAK-659 | ||
CPI-818 | ||
Crizotinib | ||
Ceritinib | ||
Copanlisib | ||
Duvelisib | Phase II ongoing | |
Tenalisib | ||
Ruxolitinib | ||
Alisertib | Phase III completed | |
Epigenetic | Valemetostat | Phase I ongoing |
Romidepsin + other (multiple) | Phase II ongoing | |
Azacitidine | Phase III ongoing | |
Romidepsin alone | Approved | |
Belinostat | ||
Other | ALRN-6924 | Phase I completed |
MCL1 inhibitors | Phase I ongoing | |
Lenalidomide | Phase II completed | |
Tipifarnib | Phase II ongoing | |
Venetoclax |
Finally, the decision regarding consolidating deep responses to targeted therapies with allogeneic stem cell transplantation is complex and should be individualized to each patient’s situation.
Conclusions
In this review, we have attempted to outline areas of progress in defining the disease biology and therapeutic targeting needed to advance the care of patients with R/R PTCL forward. Clearly, there are promising agents with tangible benefits in this patient population, yet further work is needed to: 1) refine patient selection for these therapies based on genomic, epigenomic, or immunologic tumor markers, 2) more precisely define mediators of response to these agents, and 3) understand patterns of treatment failure from multiple perspectives towards ultimately targeting these bypass mechanisms. Furthermore, future therapeutic approaches may not take the form of small molecule kinase inhibitors, but instead as stapled peptides, micro RNA-targeted therapies, enzyme degraders, enzyme agonism, or other emerging strategies. Finally, irrespective of therapeutic interventions, further work remains to gain deeper understanding into fundamental disease mechanisms to identify other rational targets for therapeutic gain.
Acknowledgments
The authors acknowledge institutional support through the NCI P30 CA008748
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Dr. Epstein-Peterson declares no conflicts of interest. He is the recipient of an AACR-AstraZeneca Lymphoma Research Fellowship and a Lacher Fellowship at MSKCC.
Dr. Horwitz has consulted, received honorarium from, or participated in advisory boards for; Acrotech Biopharma, C4 Therapeutics, Janssen, Kura Oncology, Kyowa Hakko Kirin, Myeloid Therapeutics, ONO Pharmaceuticals, Seattle Genetics, Takeda, Trillium Therapeutics, and Vividion Therapeutics. Dr. Horwitz has received research support for clinical trials from ADC Therapeutics, Affimed, Aileron, Celgene, Daiichi Sankyo, Forty Seven, Inc., Kyowa Hakko Kirin, Millennium /Takeda, Portola Pharmaceuticals, Seattle Genetics, Trillium Therapeutics, and Verastem.
References
- 1.d’Amore F, Gaulard P, Trümper L, et al. Peripheral T-cell lymphomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2015;26(suppl_5):v108–v15. [DOI] [PubMed] [Google Scholar]
- 2.Project IT-CL. International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes. Journal of clinical oncology. 2008;26(25):4124–30. [DOI] [PubMed] [Google Scholar]
- 3.Lansigan F, Horwitz SM, Pinter-Brown LC, et al. Differential outcome of patients with primary refractory vs. relapsed peripheral T-cell lymphoma: analysis from a prospective multicenter US cohort study. American Society of Hematology; Washington, DC; 2016. [Google Scholar]
- 4.Mckinney M, Moffitt AB, Gaulard P, et al. The genetic basis of hepatosplenic T-cell lymphoma. Cancer discovery. 2017;7(4):369–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Travert M, Huang Y, De Leval L, et al. Molecular features of hepatosplenic T-cell lymphoma unravels potential novel therapeutic targets. Blood. 2012;119(24):5795–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Palomero T, Couronné L, Khiabanian H, et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nature genetics. 2014;46(2):166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Odejide O, Weigert O, Lane AA, et al. A targeted mutational landscape of angioimmunoblastic T-cell lymphoma. Blood. 2014;123(9):1293–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Watatani Y, Sato Y, Miyoshi H, et al. Molecular heterogeneity in peripheral T-cell lymphoma, not otherwise specified revealed by comprehensive genetic profiling. Leukemia. 2019;33(12):2867–83. [DOI] [PubMed] [Google Scholar]
- 9.Okkenhaug K, Bilancio A, Farjot G, et al. Impaired B and T cell antigen receptor signaling in p110δ PI 3-kinase mutant mice. Science. 2002;297(5583):1031–4. [DOI] [PubMed] [Google Scholar]
- 10.Mamand S, Allchin RL, Ahearne MJ, Wagner SD. Comparison of interleukin-2-inducible kinase (ITK) inhibitors and potential for combination therapies for T-cell lymphoma. Scientific reports. 2018;8(1):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Coiffier B, Pro B, Prince HM, et al. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. Journal of Clinical Oncology. 2012;30(6):631–6. [DOI] [PubMed] [Google Scholar]
- 12.Zhang H, Lv H, Jia X, et al. Clinical significance of enhancer of zeste homolog 2 and histone deacetylases 1 and 2 expression in peripheral T‑cell lymphoma. Oncology letters. 2019;18(2):1415–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Liu J, Liang L, Huang S, et al. Aberrant differential expression of EZH2 and H3K27me3 in extranodal NK/T-cell lymphoma, nasal type, is associated with disease progression and prognosis. Human pathology. 2019;83:166–76. [DOI] [PubMed] [Google Scholar]
- 14.Ghione P, Faruque P, Mehta-Shah N, et al. T follicular helper phenotype predicts response to histone deacetylase inhibitors in relapsed/refractory peripheral T-cell lymphoma. Blood advances. 2020;4(19):4640–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Morishima S, Ishitsuka K, Izutsu K, et al. First-in-Human Study of the EZH1/2 Dual Inhibitor Valemetostat in Relapsed or Refractory Non-Hodgkin Lymphoma (NHL)-Updated Results Focusing on Adult T-Cell Leukemia-Lymphoma (ATL). American Society of Hematology; Washington, DC; 2019. [Google Scholar]
- 16.Ma A, Stratikopoulos E, Park K-S, et al. Discovery of a first-in-class EZH2 selective degrader. Nature chemical biology. 2020;16(2):214–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang D, Li W, Zhao R, et al. Stabilized peptide HDAC inhibitors derived from HDAC1 substrate H3K56 for the treatment of cancer stem–like cells in vivo. Cancer research. 2019;79(8):1769–83. [DOI] [PubMed] [Google Scholar]
- 18.Okkenhaug K, Patton DT, Bilancio A, et al. The p110δ isoform of phosphoinositide 3-kinase controls clonal expansion and differentiation of Th cells. The Journal of Immunology. 2006;177(8):5122–8. [DOI] [PubMed] [Google Scholar]
- 19.Huang D, Song TL, Nairismägi ML, et al. Evaluation of the PIK3 pathway in peripheral T‑cell lymphoma and NK/T‑cell lymphoma. British Journal of Haematology. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Horwitz SM, Koch R, Porcu P, et al. Activity of the PI3K-δ, γ inhibitor duvelisib in a phase 1 trial and preclinical models of T-cell lymphoma. Blood. 2018;131(8):888–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Huen A, Haverkos BM, Zain J, et al. Phase I/Ib Study of Tenalisib (RP6530), a Dual PI3K δ/γ Inhibitor in Patients with Relapsed/Refractory T-Cell Lymphoma. Cancers. 2020;12(8):2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pro B, Brammer JE, Casulo C, et al. Duvelisib in Patients with Relapsed/Refractory Peripheral T-Cell Lymphoma from the Phase 2 Primo Trial: Dose Optimization Efficacy Update and Expansion Phase Initial Results. American Society of Hematology: American Society of Hematology; Washington, DC; 2020. [Google Scholar]
- 23.Waldmann TA, Chen J. Disorders of the JAK/STAT pathway in T cell lymphoma pathogenesis: implications for immunotherapy. Annual review of immunology. 2017;35:533–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Koskela HL, Eldfors S, Ellonen P, et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. New England Journal of Medicine. 2012;366(20):1905–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nairismägi M, Tan J, Lim J, et al. JAK-STAT and G-protein-coupled receptor signaling pathways are frequently altered in epitheliotropic intestinal T-cell lymphoma. Leukemia. 2016;30(6):1311–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bergmann AK, Schneppenheim S, Seifert M, et al. Recurrent mutation of JAK3 in T‑cell prolymphocytic leukemia. Genes, chromosomes and cancer. 2014;53(4):309–16. [DOI] [PubMed] [Google Scholar]
- 27.Choi J, Goh G, Walradt T, et al. Genomic landscape of cutaneous T cell lymphoma. Nature genetics. 2015;47(9):1011–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Takemoto S, Mulloy JC, Cereseto A, et al. Proliferation of adult T cell leukemia/lymphoma cells is associated with the constitutive activation of JAK/STAT proteins. Proceedings of the National Academy of Sciences. 1997;94(25):13897–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Feldman AL, Vasmatzis G, Asmann YW, et al. Novel TRAF1‑ALK fusion identified by deep RNA sequencing of anaplastic large cell lymphoma. Genes, Chromosomes and Cancer. 2013;52(11):1097–102. [DOI] [PubMed] [Google Scholar]
- 30.Kataoka K, Nagata Y, Kitanaka A, et al. Integrated molecular analysis of adult T cell leukemia/lymphoma. Nature genetics. 2015;47(11):1304–15. [DOI] [PubMed] [Google Scholar]
- 31.Pogue SL, Kurosaki T, Bolen J, Herbst R. B cell antigen receptor-induced activation of Akt promotes B cell survival and is dependent on Syk kinase. The Journal of Immunology. 2000;165(3):1300–6. [DOI] [PubMed] [Google Scholar]
- 32.Chan AC, Van Oers N, Tran A, et al. Differential expression of ZAP-70 and Syk protein tyrosine kinases, and the role of this family of protein tyrosine kinases in TCR signaling. The Journal of Immunology. 1994;152(10):4758–66. [PubMed] [Google Scholar]
- 33.Feldman AL, Sun D, Law M, et al. Overexpression of Syk tyrosine kinase in peripheral T-cell lymphomas. Leukemia. 2008;22(6):1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Horwitz SM, Feldman TA, Hess BT, et al. A Phase 2 Study of the Dual SYK/JAK Inhibitor Cerdulatinib Demonstrates Good Tolerability and Clinical Response in Relapsed/Refractory Peripheral T-Cell Lymphoma and Cutaneous T-Cell Lymphoma. American Society of Hematology; Washington, DC; 2019. [Google Scholar]
- 35.Nayar R, Enos M, Prince A, et al. TCR signaling via Tec kinase ITK and interferon regulatory factor 4 (IRF4) regulates CD8+ T-cell differentiation. Proceedings of the National Academy of Sciences. 2012;109(41):E2794–E802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Liao XC, Littman DR. Altered T cell receptor signaling and disrupted T cell development in mice lacking Itk. Immunity. 1995;3(6):757–69. [DOI] [PubMed] [Google Scholar]
- 37.Boddicker RL, Razidlo GL, Dasari S, et al. Integrated mate-pair and RNA sequencing identifies novel, targetable gene fusions in peripheral T-cell lymphoma. Blood, The Journal of the American Society of Hematology. 2016;128(9):1234–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wang T, Lu Y, Polk A, et al. T-cell receptor signaling activates an ITK/NF-κB/GATA-3 axis in T-cell lymphomas facilitating resistance to chemotherapy. Clinical Cancer Research. 2017;23(10):2506–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mobasher M CPI-818, an Oral Interleukin-2-Inducible T-Cell Kinase Inhibitor. Pre-clinical Characterization and Interim Results of a Phase I/Ib Dose-Escalation Trial in Patients with Relapsed/Refractory T-Cell Lymphoma. T-Cell Lymphoma Forum, Parsippany, NJ, USA. 2020. [Google Scholar]
- 40.Mahadevan D, Spier C, Della Croce K, et al. Transcript profiling in peripheral T-cell lymphoma, not otherwise specified, and diffuse large B-cell lymphoma identifies distinct tumor profile signatures. Molecular cancer therapeutics. 2005;4(12):1867–79. [DOI] [PubMed] [Google Scholar]
- 41.Qi W, Spier C, Liu X, et al. Alisertib (MLN8237) an investigational agent suppresses Aurora A and B activity, inhibits proliferation, promotes endo-reduplication and induces apoptosis in T-NHL cell lines supporting its importance in PTCL treatment. Leukemia research. 2013;37(4):434–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.O’Connor OA, Özcan M, Jacobsen ED, et al. Randomized phase III study of alisertib or investigator’s choice (selected single agent) in patients with relapsed or refractory peripheral T-cell lymphoma. Journal of Clinical Oncology. 2019;37(8):613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gambacorti Passerini C, Farina F, Stasia A, et al. Crizotinib in advanced, chemoresistant anaplastic lymphoma kinase–positive lymphoma patients. Journal of the National Cancer Institute. 2014;106(2):djt378. [DOI] [PubMed] [Google Scholar]
- 44.Richly H, Kim TM, Schuler M, et al. Ceritinib in patients with advanced anaplastic lymphoma kinase–rearranged anaplastic large-cell lymphoma. Blood, The Journal of the American Society of Hematology. 2015;126(10):1257–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.O’Connor OA, Horwitz S, Masszi T, et al. Belinostat in patients with relapsed or refractory peripheral T-cell lymphoma: results of the pivotal phase II BELIEF (CLN-19) study. Journal of Clinical Oncology. 2015;33(23):2492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Coiffier B, Pro B, Prince HM, et al. Romidepsin for the treatment of relapsed/refractory peripheral T-cell lymphoma: pivotal study update demonstrates durable responses. Journal of hematology & oncology. 2014;7(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Horwitz SM, Moskowitz AJ, Jacobsen ED, et al. The combination of duvelisib, a PI3K-δ, γ inhibitor, and romidepsin is highly active in relapsed/refractory peripheral T-cell lymphoma with low rates of transaminitis: results of parallel multicenter, phase 1 combination studies with expansion cohorts. Blood. 2018;132(Supplement 1):683-. [Google Scholar]
- 48.Iyer SP, Neelapu SS, Burns E, et al. A Phase I/II Study to Examine the Safety and Efficacy of Pembrolizumab 200 Mg Fixed Dose Administered Every 3 Weeks (Q3W) in Combination with Romidepsin in Relapsed or Refractory Peripheral T-Cell Lymphoma (PTCL). American Society of Hematology; Washington, DC; 2019. [Google Scholar]
- 49.Amengual JE, Lichtenstein R, Lue J, et al. A phase 1 study of romidepsin and pralatrexate reveals marked activity in relapsed and refractory T-cell lymphoma. Blood, The Journal of the American Society of Hematology. 2018;131(4):397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Vu K, Wu C-H, Yang C-Y, et al. Romidepsin plus liposomal doxorubicin is safe and effective in patients with relapsed or refractory T-cell lymphoma: results of a phase I dose-escalation study. Clinical Cancer Research. 2020;26(5):1000–8. [DOI] [PubMed] [Google Scholar]
- 51.Pellegrini C, Dodero A, Chiappella A, et al. A phase II study on the role of gemcitabine plus romidepsin (GEMRO regimen) in the treatment of relapsed/refractory peripheral T-cell lymphoma patients. Journal of hematology & oncology. 2016;9(1):38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Strati P, Chihara D, Oki Y, et al. A phase I study of romidepsin and ifosfamide, carboplatin, etoposide for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma. haematologica. 2018;103(9):e416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Falchi L, Lue JK, Amengual JE, et al. A phase 1/2 study of oral 5-azacitidine and romidepsin in patients with lymphoid malignancies reveals promising activity in heavily pretreated peripheral T-cell lymphoma (PTCL). Blood. 2017;130(Supplement 1):1515-. [Google Scholar]
- 54.Reiman T, Savage KJ, Crump M, et al. A phase I study of romidepsin, gemcitabine, dexamethasone and cisplatin combination therapy in the treatment of peripheral T-cell and diffuse large B-cell lymphoma; the Canadian cancer trials group LY. 15 study. Leukemia & Lymphoma. 2019;60(4):912–9. [DOI] [PubMed] [Google Scholar]
- 55.Mehta-Shah N, Moskowitz AJ, Lunning MA, et al. A phase Ib/IIa trial of the combination of romidepsin, lenalidomide and carfilzomib in patients with relapsed/refractory lymphoma shows complete responses in relapsed and refractory B-and T-cell lymphomas. Blood. 2017;130(Supplement 1):821-. [Google Scholar]
- 56.Strati P, Nastoupil LJ, Davis RE, et al. A phase 1 trial of alisertib and romidepsin for relapsed/refractory aggressive B-cell and T-cell lymphomas. haematologica. 2020;105(1):e26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Laugesen A, Højfeldt JW, Helin K. Role of the polycomb repressive complex 2 (PRC2) in transcriptional regulation and cancer. Cold Spring Harbor perspectives in medicine. 2016;6(9):a026575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Pikman Y, Conway AS, Robichaud AL, et al. Targeting EZH2 for the treatment of hepatosplenic T-cell lymphoma. Blood Advances. 2020;4(7):1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Morschhauser F, Tilly H, Chaidos A, et al. Tazemetostat for patients with relapsed or refractory follicular lymphoma: an open-label, single-arm, multicentre, phase 2 trial. The Lancet Oncology. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dhiran K, Libien J, Brunson C, Jain P, Gupta R. EZH2 Expression Is Increased in Neoplastic T Cells as Compared With Reactive Lymphocytes and Differs in Different Subtypes of T-Cell Lymphoma. American Journal of Clinical Pathology. 2012;138(suppl_1):A161–A. [Google Scholar]
- 61.Yamagishi M, Hori M, Fujikawa D, et al. Targeting excessive EZH1 and EZH2 activities for abnormal histone methylation and transcription network in malignant lymphomas. Cell reports. 2019;29(8):2321–37. e7. [DOI] [PubMed] [Google Scholar]
- 62.Cheminant M, Bruneau J, Kosmider O, et al. Efficacy of 5-azacytidine in a TET2 mutated angioimmunoblastic T cell lymphoma. British journal of haematology. 2014;168(6):913–6. [DOI] [PubMed] [Google Scholar]
- 63.Lemonnier F, Dupuis J, Sujobert P, et al. Treatment with 5-azacytidine induces a sustained response in patients with angioimmunoblastic T-cell lymphoma. Blood. 2018;132(21):2305–9. [DOI] [PubMed] [Google Scholar]
- 64.Ruan J, Moskowitz A, Mehta-Shah N, et al. Multi-Center Phase II Study of Oral Azacitidine (CC-486) Plus CHOP As Initial Treatment for Peripheral T-Cell Lymphoma (PTCL). American Society of Hematology; Washington, DC; 2020. [Google Scholar]
- 65.Tobiasson M, Pandzic T, Cavelier L, Sander B, Wahlin BE. Angioimmunoblastic T-cell lymphoma and myelodysplastic syndrome with mutations in TET2, DNMT3 and CUX1–azacitidine induces only lymphoma remission. Leukemia & lymphoma. 2019;60(13):3316–9. [DOI] [PubMed] [Google Scholar]
- 66.Davids MS, Roberts AW, Wierda WG, et al. Long-Term Follow-up of Patients with Mantle Cell Lymphoma Treated with Venetoclax Monotherapy. Blood. 2018;132(Supplement 1):2883-. [Google Scholar]
- 67.Mato AR, Thompson M, Allan JN, et al. Real-world outcomes and management strategies for venetoclax-treated chronic lymphocytic leukemia patients in the United States. Haematologica. 2018;103(9):1511–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Spinner S, Crispatzu G, Yi J, et al. Re-activation of mitochondrial apoptosis inhibits T-cell lymphoma survival and treatment resistance. Leukemia. 2016;30(7):1520–30. [DOI] [PubMed] [Google Scholar]
- 69.Grabow S, Delbridge AR, Valente LJ, Strasser A. MCL-1 but not BCL-XL is critical for the development and sustained expansion of thymic lymphoma in p53-deficient mice. Blood, The Journal of the American Society of Hematology. 2014;124(26):3939–46. [DOI] [PubMed] [Google Scholar]
- 70.Koch R, Christie AL, Crombie JL, et al. Biomarker-driven strategy for MCL1 inhibition in T-cell lymphomas. Blood, The Journal of the American Society of Hematology. 2019;133(6):566–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Shustov AR, Horwitz SM, Zain J, et al. Preliminary results of the stapled peptide ALRN-6924, a dual inhibitor of MDMX and MDM2, in two phase IIa dose expansion cohorts in relapsed/refractory TP53 wild-type peripheral T-cell lymphoma. Blood. 2018;132(Supplement 1):1623-. [Google Scholar]
- 72.Fink EC, Ebert BL. The novel mechanism of lenalidomide activity. Blood. 2015;126(21):2366–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ramsay AG, Clear AJ, Kelly G, et al. Follicular lymphoma cells induce T-cell immunologic synapse dysfunction that can be repaired with lenalidomide: implications for the tumor microenvironment and immunotherapy. Blood, The Journal of the American Society of Hematology. 2009;114(21):4713–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Morschhauser F, Fitoussi O, Haioun C, et al. A phase 2, multicentre, single-arm, open-label study to evaluate the safety and efficacy of single-agent lenalidomide (Revlimid®) in subjects with relapsed or refractory peripheral T-cell non-Hodgkin lymphoma: The EXPECT trial. European journal of cancer. 2013;49(13):2869–76. [DOI] [PubMed] [Google Scholar]
- 75.Toumishey E, Prasad A, Dueck G, et al. Final report of a phase 2 clinical trial of lenalidomide monotherapy for patients with T‑cell lymphoma. Cancer. 2015;121(5):716–23. [DOI] [PubMed] [Google Scholar]
- 76.Witzig T, Sokol L, Kim W, et al. Tipifarnib in relapsed or refractory angioimmunoblastic T-cell lymphoma (AITL) and CXCL12+ peripheral T-cell lymphoma (PTCL): preliminary results from a phase 2 study. 2020.
- 77.Witzig TE, Sokol L, Jacobsen ED, et al. Tipifarnib in Relapsed or Refractory Angioimmunoblastic T-Cell Lymphoma (AITL) and CXCL12+ Peripheral T-Cell Lymphoma (PTCL): Preliminary Results from an Open-Label, Phase 2 Study. Blood. 2018;132(Supplement 1):2937-. [Google Scholar]
- 78.Gualberto A, Scholz C, Mishra V, Janes M, Kessler L. PS1002 RHOE, CXCL12 AND CXCR3 MAY IDENTIFY COMPLETE RESPONSES IN ACUTE MYELOID LEUKEMIA PATIENTS TREATED WITH TIPIFARNIB. HemaSphere. 2019;3(S1):450–1. [Google Scholar]
- 79.Laginestra MA, Cascione L, Motta G, et al. Whole exome sequencing reveals mutations in FAT1 tumor suppressor gene clinically impacting on peripheral T-cell lymphoma not otherwise specified. Modern Pathology. 2020;33(2):179–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Moskowitz AJ, Ghione P, Jacobsen ED, et al. Final Results of a Phase II Biomarker-Driven Study of Ruxolitinib in Relapsed and Refractory T-Cell Lymphoma. American Society of Hematology; Washington, DC; 2019. [Google Scholar]