Abstract
While all peripheral T-cell lymphomas are uncommon, certain subtypes are truly rare, with less than a few hundred cases per year in the USA. There are often no dedicated clinical trials in these rare subtypes, and data are generally limited to case reports and retrospective case series. Therefore, clinical management is often based on this limited literature and extrapolation of data from the more common, nodal T-cell lymphomas in conjunction with personal experience. Nevertheless, thanks to tremendous pre-clinical efforts to understand these rare diseases, an increasing appreciation of the biological changes that underlie these entities is forming. In this review, we attempt to summarize the relevant literature regarding the initial management of certain rare subtypes, specifically subcutaneous panniculitis-like T-cell lymphoma, hepatosplenic T-cell lymphoma, intestinal T-cell lymphomas, and extranodal NK/T-cell lymphoma. While unequivocally established approaches in these diseases do not exist, we make cautious efforts to provide our approaches to clinical management when possible.
Introduction
The mature T-cell and NK-cell lymphomas are a broad class of heterogeneous clinicopathological entities. The list of unique disease subtypes encompassed in this group is seemingly ever-growing, now numbering 34 distinct subtypes in the recently updated fifth edition of the World Health Organization (WHO) Classification of Haematolymphoid Tumours.1 Similarly, in the first edition of the International Consensus Classification (ICC) of Mature Lymphoid Neoplasms, over 30 entities are named.2 Despite the breadth of pathology, T-cell and NK-cell lymphomas are exceptionally rare diseases, with an annual incidence in the USA of under 2 cases per 100,000 persons.3 Individual disease incidence rates vary from 0.4-0.5 per 100,000 for the most common subtypes, such as peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), to ≤0.1 per 100,000 for rarer subtypes, such as hepatosplenic T-cell lymphoma (HSTCL) and extranodal NK/T-cell lymphoma (ENKL).3 This rarity has made dedicated study and clinical management of T-cell lymphomas particularly challenging, as clinicians are often forced to rely on case series and anecdotal reports in the face of relatively few clinical trials, especially for the rarest subtypes. Acknowledging this challenge, our goal herein is to summarize relevant literature on the clinical management for certain rare subtypes of mature T-cell and NK-cell lymphomas (Figure 1).
Subcutaneous panniculitis-like T-cell lymphoma
Subcutaneous panniculitis-like T-cell lymphoma (SPTCL) falls under the category of primary cutaneous T-cell lymphomas, including mycosis fungoides, which are reviewed elsewhere.4 SPTCL was first recognized as a distinct WHO entity in 2001, although until reliable immunohistochemistry markers became readily available in the early 2000s, SPTCL was often grouped with the related but distinct primary cutaneous γδ T-cell lymphoma (PCGDTCL), which demands a different therapeutic approach.5 However, it is now realized that SPTCL evolves from a mature cytotoxic αβ T cell, and cases expressing the yö T-cell receptor are considered PCGDTCL.
SPTCL classically presents in middle-aged females as multiple subcutaneous, non-ulcerated nodules or plaques.6,7 A personal or family history of autoimmunity, especially lupus erythematosus, may be present in up to one-third of patients, and the distinction between SPTCL and lupus panniculitis is often challenging.8 Involvement of the legs seems to be slightly more common, but involvement of the head/neck and trunk has been documented.6,7 Extracutaneous involvement, including nodal, bone marrow, and/or visceral disease, appears to be very uncommon and should be confirmed with biopsy. Many patients have constitutional symptoms, such as fevers and night sweats.6,7 In roughly one-fifth of patients, hemophagocytic lymphohistiocytosis (HLH) is present.7 HLH appears to be more common and more severe in individuals harboring germline mutations in HAVCR2.9-12 A suggested management approach for SPTCL in shown in Figure 2. The clinical evaluation is rooted in a proper pathological confirmation showing a lymphoid infiltrate involving fat lobules, with neoplastic cells having a mature αβ T-cell phenotype, usually CD8+ and negative for Epstein-Barr virus (EBV), with expression of cytotoxic markers. We perform staging evaluation with positron emission tomography/computed tomography (PET/CT), as most lesions are avid with a median SUV near 9.0.7 In addition, we undertake routine laboratory studies, including serology for human T-cell lymphotropic virus-1 to exclude cutaneous manifestations of adult T-cell leukemia/lymphoma. Diagnostic evaluation for HLH, such as measurement of ferritin, fibrinogen, triglycerides, and soluble IL-2 receptor, can be performed (we consider such evaluation in all patients as a baseline and always evaluate patients with high clinical suspicion). We often perform a rudimentary investigation for autoimmune disorders, particularly in patients in whom lupus panniculitis is entertained, by determining whether antinuclear antibodies and other serological markers are present. Positive or ambiguous results should prompt a formal rheumatology evaluation. Finally, we consider referring patients with a suspected or confirmed diagnosis to clinical genetics for HAVCR2 singlegene testing as a risk-stratification tool, although this testing is not widely available and not required for optimal management. No specific recommendations for additional testing of family members exist for patients with detected HAVCR2 aberrations, and we rely on genetic counseling for formal advice.
The management of SPTCL should be framed by the understanding that this is most often an indolent disorder for which immunomodulatory approaches are often effective.6,7 As noted previously, prior grouping of SPTCL with PCGDTCL - a generally very aggressive primary cutaneous lymphoma - initially led to concern that SPTCL was a similarly aggressive subtype. However, in 2008, the European Organization for Research and Treatment of Cancer compared clinical outcomes of 63 cases of SPTCL (at that time, referred to as a|3 SPTCL) versus 20 cases of PCGDTCL (at that time, referred to as yö SPTCL).6 Most patients in this series were treated with anthracycline-based combination chemotherapy, such as CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone). Five-year overall survival (OS) was markedly different between the two groups, at 82% for SPTCL versus 11% for PCGDTCL (P<0.001). A more recent, multi-institutional cohort of 95 cases of SPTCL and related adipotropic lymphoproliferative disorders between 1998 and 2018 confirmed the generally indolent behavior of SPTCL.7 With a median follow-up of 56 months, 67% of patients achieved a complete response (CR) to a median of three cumulative therapies. While relapses were common, no patients died of disease or HLH. Immunomodulatory agents included systemic steroids, cyclosporine, methotrexate, and others, with an objective response rate (ORR) of 52%. In particular, most patients treated with cyclosporine had a response (94%), and methotrexate as a first-line agent in seven patients produced a response in all seven. Therefore, for most patients, our approach is conservative immunosuppression with any of the agents above (in particular, cyclosporine or methotrexate), taking into consideration current symptoms, coexisting conditions, and concurrent medications. Other, smaller series show equally high response rates to immunosuppression.13-15 We assess response with PET/CT, and in those with evidence of HLH, with continued assessment of abnormal clinical and laboratory parameters. For those with relapsed/refractory (R/R) disease, we sequence immunosuppressive agents in efforts to avoid combination chemotherapy. In clinically aggressive or multiply relapsed disease, combination chemotherapy, often with consolidative transplant (autologous and allogeneic) has efficacy, similar to paradigms for PCGDTL or nodal peripheral T-cell lymphomas.6,7,16 The optimal management of frank HLH is unclear, and treatment of the underlying SPTCL is logical, with consideration for steroids and etoposide.17 Emerging data on ruxolitinib in the treatment of HLH, predominantly in the pediatric population, are encouraging,18 and we consider this agent in persistent HLH. At least one case report describes this approach in a patient with underlying SPTCL.19 Finally, CD30 expression is rare and, if detected, usually weak in intensity.6,7,20 Therefore, while we could consider brentuximab vedotin in multiply R/R disease, we acknowledge the absence of data.
Hepatosplenic T-cell lymphoma
HSTCL classically presents in males in the setting of chronic immune suppression or dysregulation, specifically inflammatory bowel disease (IBD) or after solid organ transplantation.21-23 In three representative case series, up to 20% of patients had IBD, an autoimmune disorder other than IBD, or had received a solid organ transplant.21-23 The association with IBD and IBD-directed therapy, in particular, has been reviewed in detail, stemming from a 2006 report of eight cases (six fatal) in seven males and one female with IBD who were treated with the tumor necrosis factor-α inhibitor, infliximab, in combination with additional immunosuppressant therapy.24 This led to a Food and Drug Administration boxed warning for infliximab in 2006. An epidemiological survey of over three million reports to the Food and Drug Administration Adverse Event Reporting System identified 30 unique cases of incident HSTCL in patients under chronic immunosuppression.25 Available evidence seems to suggest that the greatest risk for HSTCL is in those receiving concomitant tumor necrosis factor-a blockade and thiopurines. A study from the International T-cell Lymphoma Project (ITCP) shows that HSTCL can present in older individuals as well.26 HSTCL generally presents aggressively with hepatosplenomegaly and cytopenias. Bone marrow is nearly always involved, but lymphadenopathy is uncommon and should prompt consideration of a biopsy.22-24 Bone marrow or liver biopsy is mandatory to demonstrate a predominantly intrasinusoidal infiltrate of mature yδ (rarely αβ) cytotoxic T cells, commonly with isochrome 7q and trisomy 8 chromosomal abnormalities (detectable by fluorescence in situ hybridization)27-31 and mutational signatures with enrichment in genes of the JAK/STAT pathway and chromatin modification, such as SETD2.32-35 The diagnosis must be distinguished from other γδ T-cell lymphomas, particularly yδ T-cell large granular lymphocytic leukemia, through careful clinical and pathological review, focusing on histology, cytogenetics, and molecular findings if available.36 Therapy of curative intent for HSTCL entails combination chemotherapy induction followed by allogeneic hematopoietic stem cell transplant (alloHCT) in first remission. The optimal induction regimen is unclear (Table 1), although available evidence appears to suggest that CHOP is inadequate with refractory disease being common. In an early evaluation of 21 cases, all except two of whom were treated with CHOP/CHOP-like therapy, the median OS was 16 months; all patients died except the two patients who received non-CHOP induction.21 A second report of 15 cases from the MD Anderson Cancer Center similarly found that of six patients treated with CHOP/CHOP-like induction, all died within 2 years.22 In this series, all four surviving patients were treated with non-CHOP induction. An analysis of 166 cases of HSTCL between 1990 and 2018 showed a significantly increased ORR in patients receiving cytarabine/etoposide/platinum-containing regimens (ORR: 82%; CR: 56%) versus CHOP/CHOP-like regimens (ORR: 52%; CR: 38%).37 The median OS was significantly prolonged in the non-CHOP induction group at 36.5 versus 18 months (hazard ratio [HR]=0.33, 95% confidence interval [95% CI]: 0.19-0.58). In the absence of prospective or randomized data, this analysis suggests that cytarabine/etoposide/platinum-containing induction regimens are potentially superior to CHOP-based approaches and should be prioritized in curative intent strategies. Our institutional preference is ifosfamide-containing regimens, such as ICE (ifosfamide, carboplatin, etoposide), usually for two or three cycles followed by response assessment with PET/CT evaluation and bone marrow biopsy. In patients who achieve CR or near CR, we proceed quickly to alloHCT, usually with an additional cycle of therapy to minimize any break in therapy. In our published experience of 14 patients with HSTCL, most of whom were treated with ICE or IVAC (ifosfamide, etoposide, cytarabine), we observed a median OS of 59 months, with six of seven surviving patients receiving non-CHOP induction, and five of seven surviving patients receiving alloHCT.38 Most National Comprehensive Cancer Network (NCCN) centers use ICE as their initial approach although the guidelines39 also list DHAP(X) (dexamethasone, cytarabine, and a platinum), dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin), hyper-CVAD (hyperfractionated cyclophosphamide, vincristine, doxorubicin, cytarabine),22,23 and IVAC (ifosfamide etoposide, cytarabine) as other recommended regimens. European guidelines from the European Society of Medical Oncology propose ICE, IVAC, and CHOEP (CHOP plus etoposide) as induction regimens for HSTCL.40 A graft-versus-lymphoma effect is likely a critical factor in long-term survival, as most patients will relapse in the absence of alloHCT. In the analysis of 166 published cases, 2-year OS was 12% for those who did not receive any transplant versus 56% for those receiving alloHCT (although the non-transplant group likely included non-responders to therapy).37 In the largest report of alloHCT in HSTCL, estimated 3-year relapse-free survival and OS in 54 patients was 42% and 56%, respectively.41 The European Society for Bone and Marrow Transplantation made similar findings.42 While consolidation with autologous hematopoietic stem cell transplantation (autoHCT) can be considered,38 our preference, based on limited available data, is early transplant evaluation and alloHCT in eligible patients. In patients with R/R disease, we treat with alternative regimens in the absence of a clinical trial. Donor lymphocyte infusion in those who relapse after alloHCT may have effect.38
Intestinal T/NK-cell lymphomas
Enteropathy-associated T-cell lymphoma
The intestinal T-cell and NK-cell lymphomas are unified by their primary involvement of the gastrointestinal tract (Figure 3). The two most recognized entities, enteropathy-associated T-cell lymphoma (EATL, formerly referred to as type 1 EATL) and monomorphic epitheliotropic T-cell lymphoma (MEITL, formerly referred to as type II EATL) are now recognized as genomically distinct diseases.43-58 EATL is the most common intestinal T-cell lymphoma in Western countries, presenting with abdominal symptoms in individuals with a preceding or concomitant diagnosis of celiac disease. Direct visualization reveals destructive ulcerating lesions, or at times a frank mass, most commonly in the small bowel, although multifocal lesions involving other intestinal or extraintestinal sites are not uncommon.43-58 Biopsies show a pleomorphic population of medium/large-sized cells in an inflammatory background, with the neoplastic cells being most often CD4– /CD8– mature T cells, although CD4/CD8 expression is seen, frequently with 9q34 gains, 16q12 deletions, and JAK/STAT mutations.50,59-61 EATL is a recognized complication of celiac disease, although the pathogenesis is complex. EATL may be preceded by a condition known as refractory celiac disease, defined as persistent gastrointestinal symptoms and abnormal histological findings despite a strict gluten-free diet for ≥6–12 months. This etiological relationship has been reviewed by Dogan and colleagues and elsewhere (see excellent reviews and guidelines,62 recent genomic analysis and commentary,63,64 and clinical practice guidelines for refractory celiac disease from the American Gastroenterological Association65). The optimal management of EATL is undefined, and interpretation of primary literature is challenging due to the rarity of the condition, evolving classifications (previously EATL could refer to EATL, MEITL or other intestinal T-cell lymphomas), and sub-analyses of larger trials that include various histologies (Table 2). Still, essentially all reports describe an aggressive natural history with generally unsatisfactory long-term outcomes. For example, in the ITCP, 3-year progression-free survival (PFS) and OS for EATL was 28% and 30%, respectively, with a median OS of 11 months.26 The best outcomes have been observed with combination chemotherapy followed by consolidative transplantation, although the optimal induction is unclear. In the ITCP, in which nearly all patients with EATL received upfront anthracycline-based therapy, the CR rate to first-line therapy was 30%.26 Intensified efforts have varied results. In a small series of ten patients treated with six cycles of CHOEP, the response rate was again 30%.48 However, in the Nordic NLG-T-01 trial, one of the largest prospective trials in T-cell lymphoma, CHOEP-14 for six cycles (CHOP-14 in patients >60 years) followed by autoHCT in 21 patients with EATL resulted in a 5-year PFS and OS of 38% and 48%, respectively, and updated results showed a 10-year PFS and OS of 29%, highlighting potential for long-term survival with this program.66 Other studied options include a novel regimen consisting of two cycles of IVE (ifosfamide, etoposide, epirubicin) followed by two cycles of high-dose methotrexate and autoHCT.51 In a preliminary evaluation of this regimen in six patients, four achieved a CR and remained free of disease at over 1.5 years. A larger, retrospective analysis of a modified version of this regimen (later referred to as the Newcastle Regimen) tested one cycle of CHOP followed by three courses of IVE alternating with intermediate-dose methot-rexate and autoHCT in 26 patients.55 Five-year PFS and OS rates were both 68%. In comparison to a historical control of 31 patients treated with anthracycline-base therapy, there was a trend to improved CR rate with IVE/methotrexate (65% vs. 42%; P=0.06) and death was less frequent (39% vs. 81%, P=0.001). A phase II trial of this approach in T-cell lymphomas (including 11 patients with EATL) reported comparatively shorter but still encouraging survival in patients with EATL, with a 1-year PFS and OS of 45%.67 Given the frequent expression of CD30, EATL was included in the ECHELON 2 study, but only three patients were enrolled. A separate phase II study, the EATL-001 trial, evaluated BV-CHP (brentuximab vedotin, cyclophosphamide, doxorubicin and prednisolone) followed by autoHCT in 14 patients with EATL.68 A CR was observed in 64%. Three patients had primary progressive disease, but in all others, no relapses occurred, with a 2-year PFS and OS of 63% and 68%, respectively. Taken together, intensified approaches with CHOEP or BV-CHP in CD30+ tumors followed by autoHCT would be our preferred upfront approaches. Treatment of R/R disease is empiric. Second-line chemotherapy can be attempted.57 A durable response to CD30-directed chimeric antigen receptor T-cell therapy has been described.69 We assess disease response in EATL (and MEITL, see below) with PET/CT, and we consult closely with colleagues in Radiology and Gastroenterology to determine the most appropriate imaging and surveillance modality if there is difficulty in fully visualizing involved bowel regions.
Monomorphic epitheliotropic T-cell lymphoma
Like EATL, MEITL is a primary intestinal T-cell lymphoma, although there is no clear association with celiac disease and there may be a predilection in those of Asian descent.70-73 As MEITL was previously referred to as type II EATL (and before that, often grouped with intestinal T-cell lymphomas), dedicated literature is very sparse. Still, genomic studies increasingly demonstrate that MEITL is distinct from EATL.59 As the name implies, MEITL is monomorphic, usually positive for CD8 and CD56, and most commonly derived from intraepithelial γδ T cells. Cytogenetic analyses and mutational profiling are increasingly defining the genomic landscape of this disease.59,74,75
MEITL is aggressive, and while sporadic patients are included in clinical trials of patients with other peripheral T-cell lymphomas, there have been no dedicated treatment studies and the optimal approach is unclear. Patients often present with abdominal symptoms secondary to intestinal lesions, including perforation requiring upfront emergency resection.71,72 Many patients may be unable to tolerate further therapy due to debilitation. For example, in a multinational report from Asia of 38 patients, 26% did not receive any systemic therapy (all died). In this series, the median OS was 7 months, with most patients receiving anthracycline-based therapy.70 In a slightly larger series of 42 patients there was an improved median OS of 14.8 months.71 More patients in this series received chemotherapy (88%), which was predominantly CHOP (71%), with a CR rate to first-line therapy of 38%. Ability to receive chemotherapy, response to first-line therapy, and receipt of autoHCT were all significantly associated with improved OS in univariate analyses. A non-significant increase in OS was seen in seven patients who received non-CHOP induction including CHOEP, EPOCH and non-anthracycline based regimens. In the largest series to date of 71 patients, the median OS was 7.8 months and 2-year OS was only 15%.75 High frequencies of alterations and mutations in MYC, SETD2, STAT5B, and JAK3 were observed, as well as TP53 mutations in over one-third of patients. Eight patients survived beyond 2 years; all underwent surgery and six subsequently received chemotherapy. We would prioritize clinical trial enrollment in all eligible patients. In the absence of a clinical trial, we most often treat MEITL as nodal PTCL with CHOEP and plan consolidation of CR with autoHCT. However, due to the scarcity of data and the apparent higher rates of chemorefractory disease, we often restage early and change course with alternate therapy and planned alloHCT if the response is inadequate. Efforts to capitalize on aberrancies in the JAK/STAT pathway are worth considering in this aggressive disease and warrant further study.
Intestinal T-cell lymphoma, not otherwise specified
Intestinal T-cell lymphoma, NOS, introduced in the WHO-HAEM4R and listed in both the WHO1 and ICC2 classification systems, is a non-descript category for T-cell lymphomas of the intestines that do not conform to diagnostic criteria for EATL or MEITL. Whether this is a unique entity or simply represents PTCL-NOS with intestinal involvement is unclear.76 Comprehensive immunophenotyping and mutational profiling, if available, are recommended. We would most likely treat intestinal T-cell lymphoma, NOS, similarly to EATL or PTCL-NOS once confidently distinguished from the indolent processes described below.
Indolent T-cell and NK-cell disorders of the gastrointestinal tract
Indolent T-cell lymphoma of the gastrointestinal tract (referred to as indolent clonal T-cell lymphoproliferative disorder of the gastrointestinal tract in the ICC) is a clonal process marked by a chronic natural history generally without progressive, disseminated disease. Only case reports and cases series exist.77-88 Presenting symptoms include dyspepsia, vomiting, and diarrhea.86 Disease has mostly been documented in the small intestine, but involvement of the oral cavity, esophagus, and large bowel has also been reported.86 While mesenteric adenopathy can be seen, frank peripheral adenopathy or extraintestinal involvement is uncommon.86 Macroscopically, intestinal polyps may be seen, and microscopically, a non-destructive, superficial lymphoid infiltrate of mature, αβ T cells with a low proliferation rate is observed.86 Recently, recurrent STAT3-JAK2 fusions and additional mutational events resulting in JAK-STAT activation have been identified.87,88 The optimal management of indolent T-cell lymphoma of the gastrointestinal tract is unknown and has been non-uniform in the literature, but options include observation, resection, budesonide, mesalamine, interferon, and combination chemotherapy.86,88 Three tentative conclusions can be made: (i) OS is long regardless of management, including with observation, with most patients in available reports being alive, some beyond 20 years; (ii) the natural history appears to be a locally chronic disease without high risk of aggressive transformation or spread; and (iii) no therapy appears reliably curative. Based on all available literature, if symptoms need treatment, we would favor conservative therapies at first and avoid the use of chemotherapy. The related indolent NK-cell lymphoproliferative disorder of the gastrointestinal tract is a new entry in the WHO classification and also designated in the ICC, and was previously referred to as lymphomatoid gastropathy89 or NK-cell enteropathy.90 This entity was first documented in a Japanese case series in 2010, describing ten cases of a self-limited, EBV-negative NK-cell proliferation in the stomach.89 Patients were middle-aged with an equal sex distribution. Three cases were discovered upon follow-up of prior gastric malignancy, and the others were discovered via gastric cancer screening. No patients had extragastric disease, and all patients were observed and alive at the time of publication. Only three patients had recurrent disease after initial resection, and in each case recurrent lesions self-resolved without intervention. Soon after this report, an additional eight cases from the USA were published, again describing an EBV-negative NK-cell proliferation confined to the gastrointestinal tract.90 In this series, most patients presented with vague abdominal complaints. Again, no extraintestinal involvement was detected. All patients were observed endoscopically and none developed progression. No patients died. Recurrent somatic JAK3 mutations were recently identified in some cases in a series led by our center.91 This series and others92-94 all reiterate that the condition has a chronic, at times spontaneously remitting natural history for which observation may be appropriate. As in indolent T-cell lymphoma of the gastrointestinal tract, we perform full staging, including endoscopic bowel evaluation, peripheral blood flow cytometry, bone marrow assessment, and mutational profiling, with a dedicated effort to rule out PTCL or ENKL, which require treatment (see below). Close observation is recommended with endoscopic follow-up in the absence of concerning clinical or pathological features.
Extranodal NK/T-cell lymphoma
ENKL are predominantly extranodal diseases with a predilection for the upper aerodigestive tract, as first noted by Ng and colleagues over 30 years ago, recognizing a pattern of cases of sino-nasal lymphoma showing NK-cell markers.95 ENKL involving the upper aerodigestive tract has traditionally been called nasal type ENKL, whereas non-nasal ENKL or more disseminated disease with an unknown site of origin is sometimes called extranasal ENKL. The qualifier ‘nasal type’ has been retained by the ICC2 but not the WHO.1 The frequency of this disease is higher in Asian populations.96 A common presentation for nasal type ENKL is a rapidly growing, destructive mass involving any number of facial structures, including the nasopharynx or palate, with frequent invasion of local structures such as skin, sinuses, or orbits. Systemic symptoms and hemophagocytosis may be present at diagnosis. Key histopathological findings include a diffuse, aggressive lymphocytic infiltrate displaying NK-cell or T-cell markers along with universal EBER positivity. We evaluate patients with newly diagnosed, nasal type ENKL with direct visualization of the nasopharynx, CT or magnetic resonance imaging of nasal structures, PET/CT, laboratory studies including quantitative assessment of EBV DNA by polymerase chain reaction,97 and bone marrow biopsy to confirm stage. Given our nearly universal use of radiation therapy (RT) as an adjuvant or consolidation for patients with early-stage disease, we routinely consult Radiation Oncology colleagues during the initial workup for assessment of pretreatment disease volumes to facilitate planning of post-chemotherapy radiation. While we evaluate prognosis with the prognostic index of NK lymphoma plus EBV DNA scoring system (PINK-E; risk factors: age >60 years, stage III/IV, distant [non-regional] lymph node involvement, non-nasal subtype, detectable plasma EBV-DNA at diagnosis),98 our treatment decisions are primarily driven by stage of disease and the patient’s fitness, as described below.
Our treatment approach is broadly based on the stage and extent of disease, the patient’s comorbidities, candidacy for HCT, and goals of care. A number of overarching principles shape our management, including recognition of: (i) the importance of RT for local disease control; (ii) the additive benefits of systemic therapy in addition to RT, primarily with pegaspargase-based combination chemotherapy regimens to overcome inherent chemoresistance and address micrometastatic disease; (iii) the ability to consolidate response with either autoHCT or alloHCT; and (iv) the promising activity of PD-1 and PD-L1 targeting monoclonal antibodies in R/R ENKL.
For fit patients with stage I/II nasal ENKL, we typically use combined modality therapy that incorporates local RT directed at the main sites of disease plus systemic therapy. While early retrospective analyses were conflicting regarding the benefit of systemic therapy in addition to RT,99,100 later population-level data from both the International Peripheral T-Cell Lymphoma Project and the China Lymphoma Collaborative Group showed a clear survival benefit when systemic therapy is added to RT.96,101 The conflicting data from earlier reports may stem from the use of CHOP or CHOP-like chemotherapy regimens that are now recognized as suboptimal for treating ENKL. The benefits of systemic therapy in addition to RT in stage I/II ENKL were recently documented in a small, multicenter randomized control trial in China, in which 65 patients with stage I/II ENKL were randomized to RT alone versus sequential asparaginase-based chemotherapy followed by RT.102 The response rate (83.3% vs. 60.0%), 5-year PFS (82.9% vs. 56.5%), and 5-year OS (85.7% vs. 60.4%) were significantly improved with the addition of chemotherapy to RT. As expected, combined modality therapy resulted in statistically significantly increased myelosuppression, although grade III/IV events were uncommon overall. For patients ineligible for multiagent chemotherapy with stage I/II nasal ENKL, we typically use RT alone (generally at doses of 50 to 55 Gy) or combine RT with less intensive chemotherapy.
There is uncertainty regarding the optimal sequencing and type of therapy in this paradigm, and supportive data exist for sequential chemoradiation,102-104 sandwich chemoradiation,105-107 and concurrent chemoradiation (typically followed by additional chemotherapy alone).108-113 As RT alone is highly effective (especially for stage I disease)114 and concurrent chemotherapy generally compromises the choice and intensity of chemotherapy as well as adding toxicity, we typically favor sequential approaches instead, which can avoid the additive toxicity of concurrent chemoradiation and still enable early incorporation of effective systemic therapy. In our institutional practice, for fit patients, we favor one to two cycles of the modified SMILE regimen (dexamethasone, methotrexate, ifosphamide, pegaspargase, etoposide) (see full regimen descriptions in Table 3) for systemic therapy followed by RT.103 In patients with early-stage and localized nasal ENKL who attain a CR after combined modality therapy, we generally observe rather than consolidate with transplantation. Table 3 details key studies and results for different therapeutic strategies in early-stage ENKL. Dosing, radiation strategies, and even staging vary between published reports, and clinicians should consult the primary literature for specific treatment details of any particular treatment program.
For patients with advanced-stage ENKL, we use combination chemotherapy and often consider combined modality therapy in those with a symptomatic, invasive mass in the upper aerodigestive tract given the effectiveness of RT and the morbidity associated with local treatment failure. In advanced-stage disease, systemic therapy regimens all integrate L-asparaginase or pegaspargase (Table 3) and include modified SMILE,103,115,116 P-GemOx (pegaspargase, gemcitabine, oxaliplatin),117 DDGP (cisplatin, dexamethasone, gemcitabine, pegaspargase),118 and AspaMetDex (L-asparaginase, methotrexate, dexamethasone).119 At our institution, we use modified SMILE in most fit patients due to institutional experience with the regimen and its known long-term efficacy.101 Still, we acknowledge the efficacy of other asparaginase-containing regimens listed above, in particular those containing gemcitabine, which show largely comparable results with potentially less toxicity, but for which there are noticeably fewer data and less long-term follow-up in comparison to those for the SMILE regimen. Outside of China, these regimens have not been compared directly, and the most critical principle in practice is to use asparaginase-based induction and not an anthracycline-containing regimen. Of note, a randomized trial of SMILE versus DDGP was conducted in China in patients with newly diagnosed, stage III/IV ENKL.120 Patients treated with DDGP achieved improved PFS (median not reached vs. 6.8 months, HR=0.42, 95% CI: 0.23-0.77) and OS (median not reached vs. 75.2 months, HR=0.41, 95% CI: 0.19-0.89) compared to those treated with SMILE. The ORR favored the DDGP arm (90.0% vs. 60.0%, P=0.067). However, there are important caveats regarding this trial that limit its routine use in clinical practice. First, the trial applied an alternative staging system that is not routinely used in North America (called the Chinese Southwest Oncology Group and Asia Lymphoma Study Group system).121 In this system, stage III disease is any lesion with regional lymph node involvement, which would typically be considered stage II disease in the Lugano Modification of the Ann Arbor Staging System. More patients with stage III disease in this system (vs. stage IV) were in the DDGP arm, which may indicate that the DDGP arm was enriched with patients who would otherwise be considered to have early-stage disease (and likely more favorable prognoses). In addition, the SMILE regimen was significantly modified and resulted in excessive toxicity including seven treatment-related deaths (17.5% of patients treated with SMILE) and lower efficacy than in other published studies. In our own institutional experience of using the modified SMILE regimen, we did not document any treatment-related deaths,103 and we are hesitant to extrapolate the results of this Chinese trial given the non-conventional staging system used and the unusually high treatment-related mortality in the SMILE arm.
We strongly consider consolidative HCT in patients with advanced-stage disease who attain complete remission, although the decision whether to perform autoHCT or alloHCT is highly individualized based on disease presentation, chemosensitivity, treatment tolerability, patients’ fitness, and donor availability. Our practice is to consider alloHCT when possible, especially for patients with advanced-stage disease involving multiple extranodal sites, although there is no clear consensus. If the goal is alloHCT, there is no minimum number of cycles of therapy that must be performed (we proceed as fast as possible once remission has been obtained). In terms of autoHCT, a recently reported retrospective study of 20 patients who underwent upfront autoHCT who were compared to a matched, 60-patient control group who were observed suggested improved PFS and OS with autoHCT.123 There are other reports documenting the efficacy of autoHCT.123,124 In terms of alloHCT, the Center for International Blood and Marrow Transplant Research used pooled data to evaluate 82 patients with ENKL who underwent alloHCT (30 patients in first CR).125 Of note, 22% in this series were refractory to frontline therapy and only 38% received pegaspargase prior to alloHCT. With a median follow-up of 36 months, the 3-year PFS and OS was 28% and 34%, respectively. Non-relapse mortality at 3 years was 30%. The Asia Lymphoma Study Group reported outcomes for 18 patients undergoing alloHCT (9 patients in first CR), demonstrating 5-year event-free survival and OS of 51% and 57%, respectively.126 For patients with prolonged follow-up in both series, survival plateaus suggest curative potential.
Driven by EBV, PD-L1 is frequently expressed on NK/T cells as a mechanism to avert immune surveillance. For R/R ENKL, recent results on the use of anti-PD-1 and anti-PD-L1 monoclonal antibodies are encouraging (Table 4).127-133 These agents are the preferred therapies in the R/R setting, if accessible.39 Table 4 details outcomes of the various agents. None is approved in the USA and Europe, and only pembrolizumab and nivolumab are compendium-listed by the NCCN guidelines (to the best of our knowledge, none of these agents is approved globally at the time of publication).39 While pseudo-progression has been reported with these agents, hyperprogression akin to that reported in adult T-cell leukemia-lymphoma134 has not been consistently observed in ENKL. We await final results from larger series with longer follow-up in the R/R setting to assess response durability as well as the use of checkpoint blockade prior to alloHCT, given concerns regarding graft-versus-host disease. Ongoing studies are evaluating the incorporation of checkpoint blockade in first-line regimens (NCT03728972). Finally, additional EBV-directed therapies for R/R ENKL are under investigation, including nanatinostat, a selective histone deacetylase inhibitor, in combination with valganciclovir,135 and autologous EBV-specific T cells.136
A word on non-cytotoxic therapies
The last decade has witnessed numerous attempts to utilize non-cytotoxic therapies in T-cell lymphomas, primarily in the R/R setting. These agents include brentuximab vedotin,137,138 belinostat,139 romidepsin,140,141 pralatrexate,142 bortezomib,143,144 lenalidomide,145-147 duvelisib,148 and ruxolitinib,149 some of which are approved for use in the USA and Europe, and others which can be variably used off-label. The referenced clinical trials of these agents are dominated by the more common nodal T-cell lymphomas (PTCL-NOS, angioimmunoblastic T-cell lymphoma, and anaplastic large cell lymphoma), and only a handful of patients with the rare subtypes discussed in this review were included in these trials. Therefore, when referencing response rates and survival times following the use of these agents, it is imperative to realize that the true efficacy of many novel therapies remains largely unclear in the rare subtypes. An increasing reliance on molecular drivers of response and resistance may allow for a more nuanced application of therapies, as histology-specific clinical trials are challenging to conduct (although not insurmountable68) given the sheer rarity of these entities.
Conclusions
Treating rare diseases is a challenging endeavor. Informative clinical trials are lacking, and management decisions are consequently derived from imperfect data. Still, patients with rare diseases need treatment, and as clinicians we must therefore thoughtfully review prior literature, searching for themes and guiding principles that, when coupled with knowledge on underlying biology and our own personal experiences, can allow for reasonable management decisions. Thanks to tremendous basic science and translational reaearch, as well as multicenter, often international clinical efforts in the dedicated study of T-cell lymphomas, we are hopeful that the next decade can provide further insights into these rare entities and refine treatment paradigms based on greater degrees of evidence.
Footnotes
- Received May 2, 2023
- Accepted July 3, 2023
Correspondence
Disclosures
There are no disclosures related to the present manuscript. Outside of this work, RS and ZDEP have no disclosures. SMH has received research support from ADC Therapeutics, Afmed, Aileron, Celgene, Crispr Therapeutics, Daiichi Sankyo, Forty Seven, Inc., Kyowa Hakko Kirin, Millennium /Takeda, Seattle Genetics, Trillium Therapeutics, and Verastem/SecuraBio. He has also receives consultancy fees from Acrotech Biopharma, ADC Therapeutics, Astex, Auxilus Pharma, Merck, C4 Therapeutics, Celgene, Cimieo Therapeutics, Daiichi Sankyo, Janssen, Kura Oncology, Kyowa Hakko Kirin, Myeloid Therapeutics, ONO Pharmaceuticals, Seattle Genetics, SecuraBio, Shoreline Biosciences, Inc, Takeda, Trillium Therapeutics, Tubulis, Verastem/SecuraBio, Vividion Therapeutics and Yingli Pharma Limited.
Contributions
All authors wrote the paper and approved the final version of the manuscript.
References
- Alaggio R, Amador C, Anagnostopoulos I. The 5th edition of the World Health Organization classification of haematolymphoid tumours: lymphoid neoplasms. Leukemia. 2022; 36(7):1720-1748. https://doi.org/10.1038/s41375-022-01625-xPubMedPubMed CentralGoogle Scholar
- Campo E, Jaffe ES, Cook JR. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood. 2022; 140(11):1229-1253. https://doi.org/10.1182/blood.2022015851PubMedPubMed CentralGoogle Scholar
- Howlander N, Noone A, Krapcho M, Garshell J, Miller D. SEER Cancer Statistics Review. 1975-2011. Google Scholar
- Khan N, Noor SJ, Horwitz S. How we treat mycosis fungoides and Sézary syndrome. Clin Adv Hematol Oncol. 2021; 19(9):573-581. Google Scholar
- David KA, Pulitzer M, Guitart J. Characteristics, treatment patterns, and outcomes in primary cutaneous gamma delta T cell lymphoma (PCGDTCL): a real world multi-institutional analysis of a rare malignancy. Blood. 2019; 134(Suppl_1):4028. https://doi.org/10.1182/blood-2019-130223Google Scholar
- Willemze R, Jansen PM, Cerroni L. Subcutaneous panniculitis-like T-cell lymphoma: definition, classification, and prognostic factors: an EORTC Cutaneous Lymphoma Group study of 83 cases. Blood. 2008; 111(2):838-845. https://doi.org/10.1182/blood-2007-04-087288PubMedGoogle Scholar
- Guitart J, Mangold AR, Martinez-Escala ME. Clinical and pathological characteristics and outcomes among patients with subcutaneous panniculitis-like T-cell lymphoma and related adipotropic lymphoproliferative disorders. JAMA Dermatol. 2022; 158(10):1167-1174. https://doi.org/10.1001/jamadermatol.2022.3347PubMedPubMed CentralGoogle Scholar
- Rangel LK, Villa-Ruiz C, Lo K. Clinical characteristics of lupus erythematosus panniculitis/profundus: a retrospective review of 61 patients. JAMA Dermatol. 2020; 156(11):1264-1266. https://doi.org/10.1001/jamadermatol.2020.2797PubMedPubMed CentralGoogle Scholar
- Gayden T, Sepulveda FE, Khuong-Quang DA. Germline HAVCR2 mutations altering TIM-3 characterize subcutaneous panniculitis-like T cell lymphomas with hemophagocytic lymphohistiocytic syndrome. Nat Genet. 2018; 50(12):1650-1657. https://doi.org/10.1038/s41588-018-0251-4PubMedGoogle Scholar
- Polprasert C, Takeuchi Y, Kakiuchi N. Frequent germline mutations of HAVCR2 in sporadic subcutaneous panniculitis-like T-cell lymphoma. Blood Adv. 2019; 3(4):588-595. https://doi.org/10.1182/bloodadvances.2018028340PubMedPubMed CentralGoogle Scholar
- Sonigo G, Battistella M, Beylot-Barry M. HAVCR2 mutations are associated with severe hemophagocytic syndrome in subcutaneous panniculitis-like T-cell lymphoma. Blood. 2020; 135(13):1058-1061. https://doi.org/10.1182/blood.2019003811PubMedGoogle Scholar
- Koh J, Jang I, Mun S. Genetic profiles of subcutaneous panniculitis-like T-cell lymphoma and clinicopathological impact of HAVCR2 mutations. Blood Adv. 2021; 5(20):3919-3930. https://doi.org/10.1182/bloodadvances.2021004562PubMedPubMed CentralGoogle Scholar
- Guenova E, Schanz S, Hoetzenecker W. Systemic corticosteroids for subcutaneous panniculitis-like T-cell lymphoma. Br J Dermatol. 2014; 171(4):891-894. https://doi.org/10.1111/bjd.13053PubMedGoogle Scholar
- Michonneau D, Petrella T, Ortonne N. Subcutaneous panniculitis-like T-cell lymphoma: immunosuppressive drugs induce better response than polychemotherapy. Acta Derm Venereol. 2017; 97(3):358-364. https://doi.org/10.2340/00015555-2543PubMedGoogle Scholar
- López-Lerma I, Peñate Y, Gallardo F. Subcutaneous panniculitis-like T-cell lymphoma: clinical features, therapeutic approach, and outcome in a case series of 16 patients. J Am Acad Dermatol. 2018; 79(5):892-898. https://doi.org/10.1016/j.jaad.2018.05.1243PubMedGoogle Scholar
- Gibson JF, Alpdogan O, Subtil A. Hematopoietic stem cell transplantation for primary cutaneous γδ T-cell lymphoma and refractory subcutaneous panniculitis-like T-cell lymphoma. J Am Acad Dermatol. 2015; 72(6):1010-1015.e5. https://doi.org/10.1016/j.jaad.2015.01.003PubMedGoogle Scholar
- Rosée P la, Horne AC, Hines M. Recommendations for the management of hemophagocytic lymphohistiocytosis in adults. Blood. 2019; 133(23):2465-2477. https://doi.org/10.1182/blood.2018894618PubMedGoogle Scholar
- Zhang Q, Zhao YZ, Ma HH. A study of ruxolitinib responsebased stratified treatment for pediatric hemophagocytic lymphohistiocytosis. Blood. 2022; 139(24):3493-3504. https://doi.org/10.1182/blood.2021014860PubMedGoogle Scholar
- Lévy R, Fusaro M, Guerin F. Efficacy of ruxolitinib in subcutaneous panniculitis-like T-cell lymphoma and hemophagocytic lymphohistiocytosis. Blood Adv. 2020; 4(7):1383. https://doi.org/10.1182/bloodadvances.2020001497PubMedPubMed CentralGoogle Scholar
- Wehkamp U, Mitteldorf C, Stendel S. Most rare subtypes of cutaneous lymphoma display variable CD30 expression: analysis of the German Cutaneous Lymphoma Network. Br J Dermatol. 2021; 185(1):228-230. https://doi.org/10.1111/bjd.19823PubMedGoogle Scholar
- Belhadj K, Reyes F, Farcet JP. Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: report on a series of 21 patients. Blood. 2003; 102(13):4261-4269. https://doi.org/10.1182/blood-2003-05-1675PubMedGoogle Scholar
- Falchook GS, Vega F, Dang NH. Hepatosplenic gamma-delta T-cell lymphoma: clinicopathological features and treatment. Ann Oncol. 2009; 20(6):1080-1085. https://doi.org/10.1093/annonc/mdn751PubMedPubMed CentralGoogle Scholar
- Yabe M, Medeiros LJ, Tang G. Prognostic factors of hepatosplenic T-cell lymphoma: clinicopathologic study of 28 cases. Am J Surg Pathol. 2016; 40(5):676-688. https://doi.org/10.1097/PAS.0000000000000614PubMedGoogle Scholar
- Mackey AC, Green L, Liang LC, Dinndorf P, Avigan M. Hepatosplenic T cell lymphoma associated with infliximab use in young patients treated for inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2007; 44(2):265-267. https://doi.org/10.1097/MPG.0b013e31802f6424PubMedGoogle Scholar
- Deepak P, Sifuentes H, Sherid M. T-cell non-Hodgkinʼs lymphomas reported to the FDA AERS with tumor necrosis factoralpha (TNF-α) inhibitors: results of the REFURBISH study. Am J Gastroenterol. 2013; 108(1):99-105. https://doi.org/10.1038/ajg.2012.334PubMedGoogle Scholar
- Foss FM, Horwitz SM, Civallero M. Incidence and outcomes of rare T cell lymphomas from the T Cell Project: hepatosplenic, enteropathy associated and peripheral gamma delta T cell lymphomas. Am J Hematol. 2020; 95(2):151-155. https://doi.org/10.1002/ajh.25674PubMedPubMed CentralGoogle Scholar
- Wang CC, Tien HF, Lin MT. Consistent presence of isochromosome 7q in hepatosplenic T gamma/delta lymphoma: a new cytogenetic-clinicopathologic entity. Genes Chromosomes Cancer. 1995; 12(3):161-164. https://doi.org/10.1002/gcc.2870120302PubMedGoogle Scholar
- Yao M, Tien HF, Lin MT. Clinical and hematological characteristics of hepatosplenic T gamma/delta lymphoma with isochromosome for long arm of chromosome 7. Leuk Lymphoma. 1996; 22:495-500. https://doi.org/10.3109/10428199609054788PubMedGoogle Scholar
- Jonveaux P, Daniel MT, Martel V, Maarek O, Berger R. Isochromosome 7q and trisomy 8 are consistent primary, non-random chromosomal abnormalities associated with hepatosplenic T γ/δ lymphoma. Leukemia. 1996; 10(9):1453-1455. Google Scholar
- Alonsozana ELC, Stamberg J, Kumar D. Isochromosome 7q: the primary cytogenetic abnormality in hepatosplenic gammadelta T cell lymphoma. Leukemia. 1997; 11(8):1367-1372. https://doi.org/10.1038/sj.leu.2400742PubMedGoogle Scholar
- Wlodarska I, Martin-Garcia N, Achten R. Fluorescence in situ hybridization study of chromosome 7 aberrations in hepatosplenic T-cell lymphoma: isochromosome 7q as a common abnormality accumulating in forms with features of cytologic progression. Genes Chromosomes Cancer. 2002; 33(3):243-251. https://doi.org/10.1002/gcc.10021PubMedGoogle Scholar
- Travert M, Huang Y, de Leval L. Molecular features of hepatosplenic T-cell lymphoma unravels potential novel therapeutic targets. Blood. 2012; 119(24):5795-5806. https://doi.org/10.1182/blood-2011-12-396150PubMedPubMed CentralGoogle Scholar
- Ferreiro JF, Rouhigharabaei L, Urbankova H. Integrative genomic and transcriptomic analysis identified candidate genes implicated in the pathogenesis of hepatosplenic T-cell lymphoma. PLoS One. 2014; 9(7):e102977. https://doi.org/10.1371/journal.pone.0102977PubMedPubMed CentralGoogle Scholar
- Nicolae A, Xi L, Pittaluga S. Frequent STAT5B mutations in γδ hepatosplenic T-cell lymphomas. Leukemia. 2014; 28(11):2244-2248. https://doi.org/10.1038/leu.2014.200PubMedPubMed CentralGoogle Scholar
- McKinney M, Moffitt AB, Gaulard P. The genetic basis of hepatosplenic T-cell lymphoma. Cancer Discov. 2017; 7(4):369-379. https://doi.org/10.1158/2159-8290.CD-16-0330PubMedPubMed CentralGoogle Scholar
- Yabe M, Medeiros LJ, Wang SA. Distinguishing between hepatosplenic T-cell lymphoma and γδ T-cell large granular lymphocytic leukemia: a clinicopathologic, immunophenotypic, and molecular analysis. Am J Surg Pathol. 2017; 41(1):82-93. https://doi.org/10.1097/PAS.0000000000000743PubMedGoogle Scholar
- Klebaner D, Koura D, Tzachanis D. Intensive induction therapy compared with CHOP for hepatosplenic T-cell lymphoma. Clin Lymphoma Myeloma Leuk. 2020; 20(7):437. https://doi.org/10.1016/j.clml.2019.10.007PubMedGoogle Scholar
- Voss MH, Lunning MA, Maragulia JC. Intensive induction chemotherapy followed by early high-dose therapy and hematopoietic stem cell transplantation results in improved outcome for patients with hepatosplenic T-cell lymphoma: a single institution experience. Clin Lymphoma Myeloma Leuk. 2013; 13(1):8-14. https://doi.org/10.1016/j.clml.2012.09.002PubMedPubMed CentralGoogle Scholar
- Horwitz SM, Ansell S, Ai WZ. T-cell lymphomas, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022; 20(3):285-308. https://doi.org/10.6004/jnccn.2022.0015PubMedGoogle Scholar
- d’Amore F, Gaulard P, Trümper L. Peripheral T-cell lymphomas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015; 26(Suppl 5):v108-v115. https://doi.org/10.1093/annonc/mdv201PubMedGoogle Scholar
- Rashidi A, Cashen AF. Outcomes of allogeneic stem cell transplantation in hepatosplenic T-cell lymphoma. Blood Cancer J. 2015; 5(6):e318. https://doi.org/10.1038/bcj.2015.43PubMedPubMed CentralGoogle Scholar
- Tanase A, Schmitz N, Stein H. Allogeneic and autologous stem cell transplantation for hepatosplenic T-cell lymphoma: a retrospective study of the EBMT Lymphoma Working Party. Leukemia. 2015; 29(3):686-688. https://doi.org/10.1038/leu.2014.280PubMedGoogle Scholar
- Tutt ANJ, Brada M, Sampson SA. Enteropathy associated T cell lymphoma presenting as an isolated CNS lymphoma three years after diagnosis of coeliac disease: T cell receptor polymerase chain reaction studies failed to show the original enteropathy to be a clonal disorder. Gut. 1997; 40(6):801-803. https://doi.org/10.1136/gut.40.6.801PubMedPubMed CentralGoogle Scholar
- Gale J, Simmonds PD, Mead GM, Sweetenham JW, Wright DH. Enteropathy-type intestinal T-cell lymphoma: clinical features and treatment of 31 patients in a single center. J Clin Oncol. 2000; 18(4):795-803. https://doi.org/10.1200/JCO.2000.18.4.795PubMedGoogle Scholar
- Daum S, Ullrich R, Heise W. Intestinal non-Hodgkin’s lymphoma: a multicenter prospective clinical study from the German Study Group on Intestinal non-Hodgkin’s Lymphoma. J Clin Oncol. 2003; 21(14):2740-2746. https://doi.org/10.1200/JCO.2003.06.026PubMedGoogle Scholar
- Jantunen E, Juvonen E, Wiklund T, Putkonen M, Nousiainen T. High-dose therapy supported by autologous stem cell transplantation in patients with enteropathy-associated T-cell lymphoma. Leuk Lymphoma. 2003; 44(12):2163-2164. https://doi.org/10.1080/1042819031000123456PubMedGoogle Scholar
- Gobbi C, Buess M, Probst A. Enteropathy-associated T-cell lymphoma with initial manifestation in the CNS. Neurology. 2003; 60(10):1718-1719. https://doi.org/10.1212/01.WNL.0000066052.84194.6EPubMedGoogle Scholar
- Wöhrer S, Chott A, Drach J. Chemotherapy with cyclophosphamide, doxorubicin, etoposide, vincristine and prednisone (CHOEP) is not effective in patients with enteropathy-type intestinal T-cell lymphoma. Ann Oncol. 2004; 15(11):1680-1683. https://doi.org/10.1093/annonc/mdh427PubMedGoogle Scholar
- Novakovic BJ, Novakovic S, Frkovic-Grazio S. A single-center report on clinical features and treatment response in patients with intestinal T cell non-Hodgkin’s lymphomas. Oncol Rep. 2006; 16(1):191-195. https://doi.org/10.3892/or.16.1.191Google Scholar
- deLeeuw RJ, Zettl A, Klinker E. Whole-genome analysis and HLA genotyping of enteropathy-type T-cell lymphoma reveals 2 distinct lymphoma subtypes. Gastroenterology. 2007; 132(5):1902-1911. https://doi.org/10.1053/j.gastro.2007.03.036PubMedGoogle Scholar
- Bishton MJ, Haynes AP. Combination chemotherapy followed by autologous stem cell transplant for enteropathy-associated T cell lymphoma. Br J Haematol. 2007; 136(1):111-113. https://doi.org/10.1111/j.1365-2141.2006.06371.xPubMedGoogle Scholar
- Al-toma A, Verbeek WHM, Visser OJ. Disappointing outcome of autologous stem cell transplantation for enteropathy-associated T-cell lymphoma. Dig Liver Dis. 2007; 39(7):634-641. https://doi.org/10.1016/j.dld.2007.03.009PubMedGoogle Scholar
- Al-toma A, Verbeek WHM, Hadithi M, von Blomberg BME, Mulder CJJ. Survival in refractory coeliac disease and enteropathy-associated T-cell lymphoma: retrospective evaluation of single-centre experience. Gut. 2007; 56(10):1373-1378. https://doi.org/10.1136/gut.2006.114512PubMedPubMed CentralGoogle Scholar
- Verbeek WHM, van de Water JMW, Al-Toma A. Incidence of enteropathy--associated T-cell lymphoma: a nation-wide study of a population-based registry in The Netherlands. Scand J Gastroenterol. 2008; 43(11):1322-1328. https://doi.org/10.1080/00365520802240222PubMedGoogle Scholar
- Sieniawski M, Angamuthu N, Boyd K. Evaluation of enteropathy-associated T-cell lymphoma comparing standard therapies with a novel regimen including autologous stem cell transplantation. Blood. 2010; 115(18):3664-3670. https://doi.org/10.1182/blood-2009-07-231324PubMedGoogle Scholar
- Delabie J, Holte H, Vose JM. Enteropathy-associated T-cell lymphoma: clinical and histological findings from the International Peripheral T-Cell Lymphoma Project. Blood. 2011; 118(1):148-155. https://doi.org/10.1182/blood-2011-02-335216PubMedGoogle Scholar
- Raderer M, Troch M, Kiesewetter B. Second line chemotherapy in patients with enteropathy-associated T cell lymphoma: a retrospective single center analysis. Ann Hematol. 2012; 91(1):57-61. https://doi.org/10.1007/s00277-011-1236-xPubMedGoogle Scholar
- Malamut G, Chandesris O, Verkarre V. Enteropathy associated T cell lymphoma in celiac disease: a large retrospective study. Dig Liver Dis. 2013; 45(5):377-384. https://doi.org/10.1016/j.dld.2012.12.001PubMedPubMed CentralGoogle Scholar
- Tomita S, Kikuti YY, Carreras J. Genomic and immunohistochemical profiles of enteropathy-associated T-cell lymphoma in Japan. Mod Pathol. 2015; 28(10):1286-1296. https://doi.org/10.1038/modpathol.2015.85PubMedGoogle Scholar
- Nicolae A, Xi L, Pham TH. Mutations in the JAK/STAT and RAS signaling pathways are common in intestinal T-cell lymphomas. Leukemia. 2016; 30(11):2245-2247. https://doi.org/10.1038/leu.2016.178PubMedPubMed CentralGoogle Scholar
- Nairismägi ML, Tan J, Lim JQ. JAK-STAT and G-protein-coupled receptor signaling pathways are frequently altered in epitheliotropic intestinal T-cell lymphoma. Leukemia. 2016; 30(6):1311-1319. https://doi.org/10.1038/leu.2016.13PubMedPubMed CentralGoogle Scholar
- Chander U, Leeman-Neill RJ, Bhagat G. Pathogenesis of enteropathy-associated T cell lymphoma. Curr Hematol Malig Rep. 2018; 13(4):308-317. https://doi.org/10.1007/s11899-018-0459-5PubMedGoogle Scholar
- Cording S, Lhermitte L, Malamut G. Oncogenetic landscape of lymphomagenesis in coeliac disease. Gut. 2022; 71(3):497-508. https://doi.org/10.1136/gutjnl-2020-322935PubMedPubMed CentralGoogle Scholar
- Murray JA. Several faces of refractory coeliac disease type 2. Gut. 2022; 71(3):449-450. https://doi.org/10.1136/gutjnl-2021-324251PubMedGoogle Scholar
- Green PHR, Paski S, Ko CW, Rubio-Tapia A. AGA clinical practice update on management of refractory celiac disease: expert review. Gastroenterology. 2022; 163(5):1461-1469. https://doi.org/10.1053/j.gastro.2022.07.086PubMedGoogle Scholar
- Relander T, Pederson MB, Ellin F. Long-term follow-up of clinical outcome determinants and correlative biological features from the Nordic NLG-T-01 trial. Blood. 2022; 140(Suppl 1):1479-1480. https://doi.org/10.1182/blood-2022-158981Google Scholar
- Phillips EH, Lannon MM, Lopes A. High-dose chemotherapy and autologous stem cell transplantation in enteropathy-associated and other aggressive T-cell lymphomas: a UK NCRI/Cancer Research UK phase II study. Bone Marrow Transplant. 2019; 54(3):465-468. https://doi.org/10.1038/s41409-018-0294-2PubMedGoogle Scholar
- Sibon D, Khater S, Bruneau J. The EATL-001 trial: results of a phase 2 study of brentuximab vedotin and CHP followed by consolidation with high-dose therapy - autologous stem-cell transplantation (HDT-ASCT) in the frontline treatment of patients with enteropathy-associated T-cell lymphoma. Blood. 2021; 138(Suppl 1):136. https://doi.org/10.1182/blood-2021-153709Google Scholar
- Voorhees TJ, Ghosh N, Grover N. Long-term remission in multiply relapsed enteropathy-associated T-cell lymphoma following CD30 CAR T-cell therapy. Blood Adv. 2020; 4(23):5925-5928. https://doi.org/10.1182/bloodadvances.2020003218PubMedPubMed CentralGoogle Scholar
- Sun J, Lu Z, Yang D, Chen J. Primary intestinal T-cell and NK-cell lymphomas: a clinicopathological and molecular study from China focused on type II enteropathy-associated T-cell lymphoma and primary intestinal NK-cell lymphoma. Mod Pathol. 2011; 24:983-992. https://doi.org/10.1038/modpathol.2011.45PubMedGoogle Scholar
- Yi JH, Lee GW, Do YR. Multicenter retrospective analysis of the clinicopathologic features of monomorphic epitheliotropic intestinal T-cell lymphoma. Ann Hematol. 2019; 98(11):2541-2550. https://doi.org/10.1007/s00277-019-03791-yPubMedGoogle Scholar
- Tse E, Gill H, Loong F. Type II enteropathy-associated T-cell lymphoma: a multicenter analysis from the Asia Lymphoma Study Group. Am J Hematol. 2012; 87(7):663-668. https://doi.org/10.1002/ajh.23213PubMedGoogle Scholar
- Huang D, Lim JQ, Cheah DMZ. Whole-genome sequencing reveals potent therapeutic strategy for monomorphic epitheliotropic intestinal T-cell lymphoma. Blood Adv. 2020; 4(19):4769-4774. https://doi.org/10.1182/bloodadvances.2020001782PubMedPubMed CentralGoogle Scholar
- Roberti A, Dobay MP, Bisig B. Type II enteropathy-associated T-cell lymphoma features a unique genomic profile with highly recurrent SETD2 alterations. Nat Commun. 2016; 7:12602. https://doi.org/10.1038/ncomms12602PubMedPubMed CentralGoogle Scholar
- Veloza L, Cavalieri D, Missiaglia E. Monomorphic epitheliotropic intestinal T-cell lymphoma comprises morphologic and genomic heterogeneity impacting outcome. Haematologica. 2023; 108(1):181-195. https://doi.org/10.3324/haematol.2022.281226PubMedPubMed CentralGoogle Scholar
- Attygalle AD, Cabeçadas J, Gaulard P. Peripheral T-cell and NK-cell lymphomas and their mimics; taking a step forward -report on the Lymphoma Workshop of the XVIth meeting of the European Association for Haematopathology and the Society for Hematopathology. Histopathology. 2014; 64(2):171-199. https://doi.org/10.1111/his.12251PubMedPubMed CentralGoogle Scholar
- Carbonnel F, Lavergne A, Messing B. Extensive small intestinal T-cell lymphoma of low-grade malignancy associated with a new chromosomal translocation. Cancer. 1994; 73(4):1286-1291. https://doi.org/10.1002/1097-0142(19940215)73:4<1286::AID-CNCR2820730425>3.0.CO;2-9Google Scholar
- Egawa N, Fukayama M, Kawaguchi K. Relapsing oral and colonic ulcers with monoclonal T-cell infiltration. Cancer. 1995; 75(7):1728-1733. https://doi.org/10.1002/1097-0142(19950401)75:7<1728::AID-CNCR2820750727>3.0.CO;2-9Google Scholar
- Hirakawa K, Fuchigami T, Nakamura S. Primary gastrointestinal T-cell lymphoma resembling multiple lymphomatous polyposis. Gastroenterology. 1996; 111(3):778-782. https://doi.org/10.1053/gast.1996.v111.pm8780585PubMedGoogle Scholar
- Carbonnel F, D’Almagne H, Lavergne A. The clinicopathological features of extensive small intestinal CD4 T cell infiltration. Gut. 1999; 45(5):662-667. https://doi.org/10.1136/gut.45.5.662PubMedPubMed CentralGoogle Scholar
- Ranheim EA, Jones C, Zehnder JL, Warnke R, Yuen A. Spontaneously relapsing clonal, mucosal cytotoxic T-cell lymphoproliferative disorder: case report and review of the literature. Am J Surg Pathol. 2000; 24(2):296-301. https://doi.org/10.1097/00000478-200002000-00017PubMedGoogle Scholar
- Zivny J, Banner BF, Agrawal S, Pihan G, Barnard GF. CD4+ T-cell lymphoproliferative disorder of the gut clinically mimicking celiac sprue. Dig Dis Sci. 2004; 49(4):551-555. https://doi.org/10.1023/B:DDAS.0000026297.25591.62PubMedGoogle Scholar
- Isomoto H, Maeda T, Akashi T. Multiple lymphomatous polyposis of the colon originating from T-cells: a case report. Dig Liver Dis. 2004; 36(3):218-221. https://doi.org/10.1016/j.dld.2003.09.019PubMedGoogle Scholar
- Svrcek M, Garderet L, Sebbagh V. Small intestinal CD4+ T-cell lymphoma: a rare distinctive clinicopathological entity associated with prolonged survival. Virchows Archiv. 2007; 451(6):1091-1093. https://doi.org/10.1007/s00428-007-0475-7PubMedGoogle Scholar
- Margolskee E, Jobanputra V, Lewis SK. Indolent small intestinal CD4+ T-cell lymphoma is a distinct entity with unique biologic and clinical features. PLoS One. 2013; 8(7):e68343. https://doi.org/10.1371/journal.pone.0068343PubMedPubMed CentralGoogle Scholar
- Perry AM, Warnke RA, Hu Q. Indolent T-cell lymphoproliferative disease of the gastrointestinal tract. Blood. 2013; 122(22):3599-3606. https://doi.org/10.1182/blood-2013-07-512830PubMedPubMed CentralGoogle Scholar
- Sharma A, Oishi N, Boddicker RL. Recurrent STAT3-JAK2 fusions in indolent T-cell lymphoproliferative disorder of the gastrointestinal tract. Blood. 2018; 131(20):2262-2266. https://doi.org/10.1182/blood-2018-01-830968PubMedPubMed CentralGoogle Scholar
- Soderquist CR, Patel N, Murty V. Genetic and phenotypic characterization of indolent T-cell lymphoproliferative disorders of the gastrointestinal tract. Haematologica. 2020; 105(7):1895. https://doi.org/10.3324/haematol.2019.230961PubMedPubMed CentralGoogle Scholar
- Mansoor A, Pittaluga S, Beck PL. NK-cell enteropathy: a benign NK-cell lymphoproliferative disease mimicking intestinal lymphoma: clinicopathologic features and follow-up in a unique case series. Blood. 2011; 117(5):1447. https://doi.org/10.1182/blood-2010-08-302737PubMedPubMed CentralGoogle Scholar
- Takeuchi K, Yokoyama M, Ishizawa S. Lymphomatoid gastropathy: a distinct clinicopathologic entity of self-limited pseudomalignant NK-cell proliferation. Blood. 2010; 116(25):5631-5637. https://doi.org/10.1182/blood-2010-06-290650PubMedGoogle Scholar
- Xiao W, Gupta GK, Yao J. Recurrent somatic JAK3 mutations in NK-cell enteropathy. Blood. 2019; 134(12):986-991. https://doi.org/10.1182/blood.2019001443PubMedPubMed CentralGoogle Scholar
- McElroy MK, Read WL, Harmon GS, Weidner N. A unique case of an indolent CD56-positive T-cell lymphoproliferative disorder of the gastrointestinal tract: a lesion potentially misdiagnosed as natural killer/T-cell lymphoma. Ann Diagn Pathol. 2011; 15(5):370-375. https://doi.org/10.1016/j.anndiagpath.2010.05.007PubMedGoogle Scholar
- Isom JA, Arroyo MR, Reddy D. NK cell enteropathy: a case report with 10 years of indolent clinical behaviour. Histopathology. 2018; 73(2):345-350. https://doi.org/10.1111/his.13502PubMedGoogle Scholar
- Xia D, Morgan EA, Berger D. NK-cell enteropathy and similar indolent lymphoproliferative disorders. Am J Clin Pathol. 2019; 151(1):75-85. https://doi.org/10.1093/ajcp/aqy108PubMedGoogle Scholar
- Chan JKC, Lo STH. Expression of natural killer cell markers in non-Hodgkin’s lymphomas. Hum Pathol. 1987; 18(12):1257-1262. https://doi.org/10.1016/S0046-8177(87)80410-0PubMedGoogle Scholar
- Au WY, Weisenburger DD, Intragumtornchai T. Clinical differences between nasal and extranasal natural killer/T-cell lymphoma: a study of 136 cases from the International Peripheral T-Cell Lymphoma Project. Blood. 2009; 113(17):3931-3937. https://doi.org/10.1182/blood-2008-10-185256PubMedGoogle Scholar
- Au WY, Pang A, Choy C, Chim CS, Kwong YL. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004; 104(1):243-249. https://doi.org/10.1182/blood-2003-12-4197PubMedGoogle Scholar
- Kim SJ, Yoon DH, Jaccard A. A prognostic index for natural killer cell lymphoma after non-anthracycline-based treatment: a multicentre, retrospective analysis. Lancet Oncol. 2016; 17(3):389-400. https://doi.org/10.1016/S1470-2045(15)00533-1PubMedGoogle Scholar
- Cheung MMC, Chan JKC, Lau WH, Ngan RKC, Foo WWL. Early stage nasal NK/T-cell lymphoma: clinical outcome, prognostic factors, and the effect of treatment modality. Int J Radiat Oncol Biol Phys. 2002; 54(1):182-190. https://doi.org/10.1016/S0360-3016(02)02916-4PubMedGoogle Scholar
- Kim K, Chie EK, Kim CW, Kim IH, Park CI. Treatment outcome of angiocentric T-cell and NK/T-cell lymphoma, nasal type: radiotherapy versus chemoradiotherapy. Jpn J Clin Oncol. 2005; 35(1):1-5. https://doi.org/10.1093/jjco/hyi006PubMedGoogle Scholar
- Deng XW, Wu JX, Wu T. Radiotherapy is essential after complete response to asparaginase-containing chemotherapy in early-stage extranodal nasal-type NK/T-cell lymphoma: a multicenter study from the China Lymphoma Collaborative Group (CLCG). Radiother Oncol. 2018; 129(1):3-9. https://doi.org/10.1016/j.radonc.2018.04.026PubMedGoogle Scholar
- Zhang L, Wang Y, Li X. Radiotherapy vs sequential pegaspargase, gemcitabine, cisplatin and dexamethasone and radiotherapy in newly diagnosed early natural killer/T-cell lymphoma: a randomized, controlled, open-label, multicenter study. Int J Cancer. 2021; 148(6):1470-1477. https://doi.org/10.1002/ijc.33329PubMedGoogle Scholar
- Ghione P, Qi S, Imber BS. Modified SMILE (mSMILE) and intensity-modulated radiotherapy (IMRT) for extranodal NK-T lymphoma nasal type in a single-center population. Leuk Lymphoma. 2020; 61(14):3331-3341. https://doi.org/10.1080/10428194.2020.1811864PubMedPubMed CentralGoogle Scholar
- Zhang Y, Ma S, Cai J. Sequential P-GEMOX and radiotherapy for early-stage extranodal natural killer/T-cell lymphoma: a multicenter study. Am J Hematol. 2021; 96(11):1481-1490. https://doi.org/10.1002/ajh.26335PubMedPubMed CentralGoogle Scholar
- Wang L, Wang ZH, Chen XQ. First-line combination of GELOX followed by radiation therapy for patients with stage IE/IIE ENKTL: an updated analysis with long-term follow-up. Oncol Lett. 2015; 10(2):1036. https://doi.org/10.3892/ol.2015.3327PubMedPubMed CentralGoogle Scholar
- Zhang L, Jiang M, Xie L. Five-year analysis from phase 2 trial of “sandwich” chemoradiotherapy in newly diagnosed, stage IE to IIE, nasal type, extranodal natural killer/T-cell lymphoma. Cancer Med. 2016; 5(1):33-40. https://doi.org/10.1002/cam4.569PubMedPubMed CentralGoogle Scholar
- Jing X-M, Zhang Z-H, Wu P. Efficacy and tolerance of pegaspargase, gemcitabine and oxaliplatin with sandwiched radiotherapy in the treatment of newly-diagnosed extranodal nature killer (NK)/T cell lymphoma. Leuk Res. 2016; 47:26-31. https://doi.org/10.1016/j.leukres.2016.05.004PubMedGoogle Scholar
- Kim SJ, Kim K, Kim BS. Phase II trial of concurrent radiation and weekly cisplatin followed by VIPD chemotherapy in newly diagnosed, stage IE to IIE, nasal, extranodal NK/T-cell lymphoma: Consortium for Improving Survival of Lymphoma study. J Clin Oncol. 2009; 27(35):6027-6032. https://doi.org/10.1200/JCO.2009.23.8592PubMedGoogle Scholar
- Yamaguchi M, Tobinai K, Oguchi M. Phase I/II study of concurrent chemoradiotherapy for localized nasal natural killer/T-cell lymphoma: Japan Clinical Oncology Group study JCOG0211. J Clin Oncol. 2009; 27(33):5594-5600. https://doi.org/10.1200/JCO.2009.23.8295PubMedGoogle Scholar
- Yamaguchi M, Tobinai K, Oguchi M. Concurrent chemoradiotherapy for localized nasal natural killer/T-cell lymphoma: an updated analysis of the Japan Clinical Oncology Group study JCOG0211. J Clin Oncol. 2012; 30(32):4044-4046. https://doi.org/10.1200/JCO.2012.45.6541PubMedGoogle Scholar
- Yamaguchi M, Suzuki R, Oguchi M. Treatments and outcomes of patients with extranodal natural killer/T-cell lymphoma diagnosed between 2000 and 2013: a cooperative study in Japan. J Clin Oncol. 2017; 35(1):32-39. https://doi.org/10.1200/JCO.2016.68.1619PubMedGoogle Scholar
- Tsai HJ, Lin SF, Chen CC. Long-term results of a phase II trial with frontline concurrent chemoradiotherapy followed by consolidation chemotherapy for localized nasal natural killer/T-cell lymphoma. Eur J Haematol. 2015; 94(2):130-137. https://doi.org/10.1111/ejh.12405PubMedGoogle Scholar
- Oh D, Ahn YC, Kim SJ, Kim WS, Ko YH. Concurrent chemoradiation therapy followed by consolidation chemotherapy for localized extranodal natural killer/T-cell lymphoma, nasal type. Int J Radiat Oncol Biol Phys. 2015; 93(3):677-683. https://doi.org/10.1016/j.ijrobp.2015.07.2267PubMedGoogle Scholar
- Li YX, Wang H, Jin J. Radiotherapy alone with curative intent in patients with stage I extranodal nasal-type NK/T-cell lymphoma. Int J Radiat Oncol Biol Phys. 2012; 82(5):1809-1815. https://doi.org/10.1016/j.ijrobp.2010.10.040PubMedGoogle Scholar
- Yamaguchi M, Kwong YL, Kim WS. Phase II study of SMILE chemotherapy for newly diagnosed stage IV, relapsed, or refractory extranodal natural killer (NK)/T-cell lymphoma, nasal type: the NK-Cell Tumor Study Group study. J Clin Oncol. 2011; 29(33):4410-4416. https://doi.org/10.1200/JCO.2011.35.6287PubMedGoogle Scholar
- Kwong YL, Kim WS, Lim ST. SMILE for natural killer/T-cell lymphoma: analysis of safety and efficacy from the Asia Lymphoma Study Group. Blood. 2012; 120(15):2973-2980. https://doi.org/10.1182/blood-2012-05-431460PubMedGoogle Scholar
- Wang JH, Wang L, Liu CC. Efficacy of combined gemcitabine, oxaliplatin and pegaspargase (P-gemox regimen) in patients with newly diagnosed advanced-stage or relapsed/refractory extranodal NK/T-cell lymphoma. Oncotarget. 2016; 7(20):29092-29101. https://doi.org/10.18632/oncotarget.8647PubMedPubMed CentralGoogle Scholar
- Zhao Q, Fan S, Chang Y. Clinical efficacy of cisplatin, dexamethasone, gemcitabine and pegaspargase (DDGP) in the initial treatment of advanced stage (stage III–IV) extranodal NK/T-cell lymphoma, and its correlation with Epstein-Barr virus. Cancer Manag Res. 2019; 11:3564. https://doi.org/10.2147/CMAR.S191929PubMedPubMed CentralGoogle Scholar
- Jaccard A, Gachard N, Marin B. Efficacy of L-asparaginase with methotrexate and dexamethasone (AspaMetDex regimen) in patients with refractory or relapsing extranodal NK/T-cell lymphoma, a phase 2 study. Blood. 2011; 117(6):1834-1839. https://doi.org/10.1182/blood-2010-09-307454PubMedGoogle Scholar
- Wang X, Zhang L, Liu X. Efficacy and safety of a pegasparaginase-based chemotherapy regimen vs an L-asparaginase-based chemotherapy regimen for newly diagnosed advanced extranodal natural killer/T-cell lymphoma: a randomized clinical trial. JAMA Oncol. 2022; 8(7):1035-1041. https://doi.org/10.1001/jamaoncol.2022.1968PubMedPubMed CentralGoogle Scholar
- Hong H, Li Y, Lim ST. A proposal for a new staging system for extranodal natural killer T-cell lymphoma: a multicenter study from China and Asia Lymphoma Study Group. Leukemia. 2020; 34(8):2243-2248. https://doi.org/10.1038/s41375-020-0740-1PubMedPubMed CentralGoogle Scholar
- Wang J, Wei L, Ye J. Autologous hematopoietic stem cell transplantation may improve long-term outcomes in patients with newly diagnosed extranodal natural killer/T-cell lymphoma, nasal type: a retrospective controlled study in a single center. Int J Hematol. 2018; 107(1):98-104. https://doi.org/10.1007/s12185-017-2324-zPubMedGoogle Scholar
- Lee J, Au WY, Park MJ. Autologous hematopoietic stem cell transplantation in extranodal natural killer/T cell lymphoma: a multinational, multicenter, matched controlled study. Biol Blood Marrow Transplant. 2008; 14(12):1356-1364. https://doi.org/10.1016/j.bbmt.2008.09.014PubMedGoogle Scholar
- Yhim HY, Kim JS, Mun YC. Clinical outcomes and prognostic factors of up-front autologous stem cell transplantation in patients with extranodal natural killer/T cell lymphoma. Biol Blood Marrow Transplant. 2015; 21(9):1597-1604. https://doi.org/10.1016/j.bbmt.2015.05.003PubMedGoogle Scholar
- Kanate AS, DiGilio A, Ahn KW. Allogeneic haematopoietic cell transplantation for extranodal natural killer/T-cell lymphoma, nasal type: a CIBMTR analysis. Br J Haematol. 2018; 182(6):916-920. https://doi.org/10.1111/bjh.14879PubMedPubMed CentralGoogle Scholar
- Tse E, Chan TSY, Koh LP. Allogeneic haematopoietic SCT for natural killer/T-cell lymphoma: a multicentre analysis from the Asia Lymphoma Study Group. Bone Marrow Transplant. 2014; 49(7):902-906. https://doi.org/10.1038/bmt.2014.65PubMedGoogle Scholar
- Kwong YL, Chan TSY, Tan D. PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing l-asparaginase. Blood. 2017; 129(17):2437-2442. https://doi.org/10.1182/blood-2016-12-756841PubMedGoogle Scholar
- Chan TSY, Li J, Loong F. PD1 blockade with low-dose nivolumab in NK/T cell lymphoma failing L-asparaginase: efficacy and safety. Ann Hematol. 2018; 97(1):193-196. https://doi.org/10.1007/s00277-017-3127-2PubMedGoogle Scholar
- Huang H, Tao R, Hao S. Sugemalimab monotherapy for patients with relapsed or refractory extranodal natural killer/T-cell lymphoma (GEMSTONE-201): results from a single-arm, multicenter, phase II study. J Clin Oncol. 2023; 41(16):3032-3041. https://doi.org/10.1200/JCO.22.02367PubMedPubMed CentralGoogle Scholar
- Huang H, Tao R, Zou L. Preliminary results from a multicenter, single-arm, phase 2 study of CS1001, an anti-programmed death-ligand 1 (PD-L1) human monoclonal antibody (mAb), in patients (pts) with relapsed or refractory extranodal natural killer/T cell lymphoma (rr-ENKTL). Blood. 2019; 134(Suppl_1):2833. https://doi.org/10.1182/blood-2019-121865Google Scholar
- Kim SJ, Lim JQ, Laurensia Y. Avelumab for the treatment of relapsed or refractory extranodal NK/T-cell lymphoma: an open-label phase 2 study. Blood. 2020; 136(24):2754-2763. https://doi.org/10.1182/blood.2020007247PubMedGoogle Scholar
- Bachy E, Savage KJ, Huang H. Treating relapsed/refractory mature T- and NK-cell neoplasms with tislelizumab: a multicenter open label phase 2 study. Blood Adv. 2023; 7(16):4435-4447. https://doi.org/10.1182/bloodadvances.2022009575PubMedPubMed CentralGoogle Scholar
- Tao R, Fan L, Song Y. Sintilimab for relapsed/refractory extranodal NK/T cell lymphoma: a multicenter, single-arm, phase 2 trial (ORIENT-4). Signal Transduct Target Ther. 2021; 6(1):365. https://doi.org/10.1038/s41392-021-00768-0PubMedPubMed CentralGoogle Scholar
- Ratner L, Waldmann TA, Janakiram M, Brammer JE. Rapid progression of adult T-cell leukemia–lymphoma after PD-1 inhibitor therapy. N Engl J Med. 2018; 378(20):1947-1948. https://doi.org/10.1056/NEJMc1803181PubMedGoogle Scholar
- Haverkos BM, Alpdogan O, Baiocchi R. Nanatinostat (Nstat) and valganciclovir (VGCV) in relapsed/refractory (R/R) Epstein-Barr virus-positive (EBV +) lymphomas: final results from the phase 1b/2 VT3996-201 study. Blood. 2021; 138(Suppl 1):623. https://doi.org/10.1182/blood-2021-152603Google Scholar
- Kim WS, Oki Y, Kim SJ. Autologous EBV-specific T cell treatment results in sustained responses in patients with advanced extranodal NK/T lymphoma: results of a multicenter study. Ann Hematol. 2021; 100(10):2529-2539. https://doi.org/10.1007/s00277-021-04558-0PubMedGoogle Scholar
- Pro B, Advani R, Brice P. Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. J Clin Oncol. 2012; 30(18):2190-2196. https://doi.org/10.1200/JCO.2011.38.0402PubMedGoogle Scholar
- Horwitz SM, Advani RH, Bartlett NL. Objective responses in relapsed T-cell lymphomas with single-agent brentuximab vedotin. Blood. 2014; 123(20):3095-3100. https://doi.org/10.1182/blood-2013-12-542142PubMedPubMed CentralGoogle Scholar
- O’Connor OA, Horwitz S, Masszi T. Belinostat in patients with relapsed or refractory peripheral T-cell lymphoma: results of the pivotal phase II BELIEF (CLN-19) study. J Clin Oncol. 2015; 33(23):2492-2499. https://doi.org/10.1200/JCO.2014.59.2782PubMedPubMed CentralGoogle Scholar
- Piekarz RL, Frye R, Prince HM. Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma. Blood. 2011; 117(22):5827-5834. https://doi.org/10.1182/blood-2010-10-312603PubMedPubMed CentralGoogle Scholar
- Coiffier B, Pro B, Prince HM. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J Clin Oncol. 2012; 30(6):631-636. https://doi.org/10.1200/JCO.2011.37.4223PubMedGoogle Scholar
- O’Connor OA, Pro B, Pinter-Brown L. Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol. 2011; 29(9):1182-1189. https://doi.org/10.1200/JCO.2010.29.9024PubMedPubMed CentralGoogle Scholar
- Evens AM, Rosen ST, Helenowski I. A phase I/II trial of bortezomib combined concurrently with gemcitabine for relapsed or refractory DLBCL and peripheral T-cell lymphomas. Br J Haematol. 2013; 163(1):55-61. https://doi.org/10.1111/bjh.12488PubMedGoogle Scholar
- Tan D, Phipps C, Hwang WYK. Panobinostat in combination with bortezomib in patients with relapsed or refractory peripheral T-cell lymphoma: an open-label, multicentre phase 2 trial. Lancet Haematol. 2015; 2(8):e326-333. https://doi.org/10.1016/S2352-3026(15)00097-6PubMedGoogle Scholar
- Zinzani PL, Pellegrini C, Broccoli A. Lenalidomide monotherapy for relapsed/refractory peripheral T-cell lymphoma not otherwise specified. Leuk Lymphoma. 2011; 52(8):1585-1588. https://doi.org/10.3109/10428194.2011.573031PubMedGoogle Scholar
- Morschhauser F, Fitoussi O, Haioun C. 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. Eur J Cancer. 2013; 49(13):2869-2876. https://doi.org/10.1016/j.ejca.2013.04.029PubMedGoogle Scholar
- Toumishey E, Prasad A, Dueck G. Final report of a phase 2 clinical trial of lenalidomide monotherapy for patients with T-cell lymphoma. Cancer. 2015; 121(5):716-723. https://doi.org/10.1002/cncr.29103PubMedGoogle Scholar
- Zinzani PL, Zain J, Mead M. P1172: duvelisib in patients with relapsed/refractory peripheral T-cell lymphoma from the phase 2 PRIMO trial: updated expansion phase analysis. Hemasphere. 2022; 6:1058-1059. https://doi.org/10.1097/01.HS9.0000847552.42271.7cPubMed CentralGoogle Scholar
- Moskowitz AJ, Ghione P, Jacobsen E. A phase 2 biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas. Blood. 2021; 138(26):2828-2837. https://doi.org/10.1182/blood.2021013379PubMedPubMed CentralGoogle Scholar
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.