NCCN Guidelines Insights: B-Cell Lymphomas, Version 3.2019

Featured Updates to the NCCN Guidelines

Authors:
Andrew D. ZelenetzMemorial Sloan Kettering Cancer Center;

Search for other papers by Andrew D. Zelenetz in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Leo I. GordonRobert H. Lurie Comprehensive Cancer Center of Northwestern University;

Search for other papers by Leo I. Gordon in
Current site
Google Scholar
PubMed
Close
 MD
,
Jeremy S. AbramsonMassachusetts General Hospital Cancer Center;

Search for other papers by Jeremy S. Abramson in
Current site
Google Scholar
PubMed
Close
 MD
,
Ranjana H. AdvaniStanford Cancer Institute;

Search for other papers by Ranjana H. Advani in
Current site
Google Scholar
PubMed
Close
 MD
,
Nancy L. BartlettSiteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine;

Search for other papers by Nancy L. Bartlett in
Current site
Google Scholar
PubMed
Close
 MD
,
Paolo F. CaimiCase Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute;

Search for other papers by Paolo F. Caimi in
Current site
Google Scholar
PubMed
Close
 MD
,
Julie E. ChangUniversity of Wisconsin Carbone Cancer Center;

Search for other papers by Julie E. Chang in
Current site
Google Scholar
PubMed
Close
 MD
,
Julio C. ChavezMoffitt Cancer Center;

Search for other papers by Julio C. Chavez in
Current site
Google Scholar
PubMed
Close
 MD
,
Beth ChristianThe Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute;

Search for other papers by Beth Christian in
Current site
Google Scholar
PubMed
Close
 MD
,
Luis E. FayadThe University of Texas MD Anderson Cancer Center;

Search for other papers by Luis E. Fayad in
Current site
Google Scholar
PubMed
Close
 MD
,
Martha J. GlennHuntsman Cancer Institute at the University of Utah;

Search for other papers by Martha J. Glenn in
Current site
Google Scholar
PubMed
Close
 MD
,
Thomas M. HabermannMayo Clinic Cancer Center;

Search for other papers by Thomas M. Habermann in
Current site
Google Scholar
PubMed
Close
 MD
,
Nancy Lee HarrisMassachusetts General Hospital Cancer Center;

Search for other papers by Nancy Lee Harris in
Current site
Google Scholar
PubMed
Close
 MD
,
Francisco Hernandez-IlizaliturriRoswell Park Comprehensive Cancer Center;

Search for other papers by Francisco Hernandez-Ilizaliturri in
Current site
Google Scholar
PubMed
Close
 MD
,
Mark S. KaminskiUniversity of Michigan Rogel Cancer Center;

Search for other papers by Mark S. Kaminski in
Current site
Google Scholar
PubMed
Close
 MD
,
Christopher R. KelseyDuke Cancer Institute;

Search for other papers by Christopher R. Kelsey in
Current site
Google Scholar
PubMed
Close
 MD
,
Nadia KhanFox Chase Cancer Center;

Search for other papers by Nadia Khan in
Current site
Google Scholar
PubMed
Close
 MD
,
Susan KrivacicConsultant;

Search for other papers by Susan Krivacic in
Current site
Google Scholar
PubMed
Close
 MPAff
,
Ann S. LaCasceDana-Farber/Brigham and Women’s Cancer Center;

Search for other papers by Ann S. LaCasce in
Current site
Google Scholar
PubMed
Close
 MD
,
Amitkumar MehtaO’Neal Comprehensive Cancer Center at UAB;

Search for other papers by Amitkumar Mehta in
Current site
Google Scholar
PubMed
Close
 MD
,
Auayporn NademaneeCity of Hope National Medical Center;

Search for other papers by Auayporn Nademanee in
Current site
Google Scholar
PubMed
Close
 MD
,
Rachel RabinovitchUniversity of Colorado Cancer Center;

Search for other papers by Rachel Rabinovitch in
Current site
Google Scholar
PubMed
Close
 MD
,
Nishitha ReddyVanderbilt-Ingram Cancer Center;

Search for other papers by Nishitha Reddy in
Current site
Google Scholar
PubMed
Close
 MD
,
Erin ReidUC San Diego Moores Cancer Center;

Search for other papers by Erin Reid in
Current site
Google Scholar
PubMed
Close
 MD
,
Kenneth B. RobertsYale Cancer Center/Smilow Cancer Hospital;

Search for other papers by Kenneth B. Roberts in
Current site
Google Scholar
PubMed
Close
 MD
,
Stephen D. SmithUniversity of Washington/Seattle Cancer Care Alliance;

Search for other papers by Stephen D. Smith in
Current site
Google Scholar
PubMed
Close
 MD
,
Erin D. SnyderO’Neal Comprehensive Cancer Center at UAB;

Search for other papers by Erin D. Snyder in
Current site
Google Scholar
PubMed
Close
 MD
,
Lode J. SwinnenThe Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins;

Search for other papers by Lode J. Swinnen in
Current site
Google Scholar
PubMed
Close
 MB, ChB
,
Julie M. VoseFred & Pamela Buffett Cancer Center; and

Search for other papers by Julie M. Vose in
Current site
Google Scholar
PubMed
Close
 MD, MBA
,
Mary A. DwyerNational Comprehensive Cancer Network.

Search for other papers by Mary A. Dwyer in
Current site
Google Scholar
PubMed
Close
 MS, CGC
, and
Hema SundarNational Comprehensive Cancer Network.

Search for other papers by Hema Sundar in
Current site
Google Scholar
PubMed
Close
 PhD
Full access

Diffuse large B-cell lymphomas (DLBCLs) and follicular lymphoma (FL) are the most common subtypes of B-cell non-Hodgkin’s lymphomas in adults. Histologic transformation of FL to DLBCL (TFL) occurs in approximately 15% of patients and is generally associated with a poor clinical outcome. Phosphatidylinositol 3-kinase (PI3K) inhibitors have shown promising results in the treatment of relapsed/refractory FL. CAR T-cell therapy (axicabtagene ciloleucel and tisagenlecleucel) has emerged as a novel treatment option for relapsed/refractory DLBCL and TFL. These NCCN Guidelines Insights highlight important updates to the NCCN Guidelines for B-Cell Lymphomas regarding the treatment of TFL and relapsed/refractory FL and DLBCL.

NCCN: Continuing Education

Target Audience: This activity is designed to meet the educational needs of physicians, nurses, pharmacists, and other healthcare professionals who manage patients with cancer.

Accreditation Statements

In support of improving patient care, National Comprehensive Cancer Network (NCCN) is jointly accredited by the Accreditation Council for Continuing Medical Education (ACCME), the Accreditation Council for Pharmacy Education (ACPE), and the American Nurses Credentialing Center (ANCC), to provide continuing education for the healthcare team.

Medicine (ACCME): NCCN designates this journal-based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

Nursing (ANCC): NCCN designates this educational activity for a maximum of 1.0 contact hour.

Pharmacy (ACPE): NCCN designates this knowledge-based continuing education activity for 1.0 contact hour (0.1 CEUs) of continuing education credit. UAN: JA4008196-0000-19-008-H01-P

All clinicians completing this activity will be issued a certificate of participation. To participate in this journal CE activity: (1) review the educational content; (2) take the posttest with a 66% minimum passing score and complete the evaluation at https://education.nccn.org/node/85881; and (3) view/print certificate.

Pharmacists: You must complete the posttest and evaluation within 30 days of the activity. Continuing pharmacy education credit is reported to the CPE Monitor once you have completed the posttest and evaluation and claimed your credits. Before completing these requirements, be sure your NCCN profile has been updated with your NAPB e-profile ID and date of birth. Your credit cannot be reported without this information. If you have any questions, please e-mail education@nccn.org.

Release date: June 10, 2019; Expiration date: June 10, 2020

Learning Objectives:

Upon completion of this activity, participants will be able to:

  • Integrate into professional practice the updates to the NCCN Guidelines for B-Cell Lymphomas

  • Describe the rationale behind the decision-making process for developing the NCCN Guidelines for B-Cell Lymphomas

Disclosure of Relevant Financial Relationships

The NCCN staff listed below discloses no relevant financial relationships:

Kerrin M. Rosenthal, MA; Kimberly Callan, MS; Genevieve Emberger Hartzman, MA; Erin Hesler; Kristina M. Gregory, RN, MSN, OCN; Rashmi Kumar, PhD; Karen Kanefield; and Kathy Smith.

Individuals Who Provided Content Development and/or Authorship Assistance:

Andrew D. Zelenetz, MD, PhD, Panel Chair, has disclosed that he receives consulting fees/honoraria from Amgen Inc.; Celgene Corporation; Gilead Sciences, Inc.; Janssen Pharmaceutica Products, LP; Novartis Pharmaceuticals Corporation; Adaptive Biotechnologies Corporation; Genentech/Roche; AbbVie, Inc.; AstraZeneca Pharmaceuticals LP; Pharmacyclics; and MorphoSys AG. He also receives grant/research support from BeiGene, Gilead Sciences, Inc., MEI Pharma Inc., and Roche Laboratories; receives an other financial benefit from BeiGene; and is a scientific advisor for AbbVie Inc., and AstraZeneca Pharmaceuticals LP.

Leo I. Gordon, MD, Vice Chair, has disclosed that he is a scientific advisor for Juno Therapeutics/Celgene Corporation, Gilead Sciences, Inc., and Bayer HealthCare; receives grant/research support from Juno Therapeutics/Celgene Corporation; and has ownership/partnership/principal in Zylem.

Jeremy S. Abramson, MD, Panel Member, has disclosed that he receives consulting fees/honoraria from AbbVie Inc., Bayer, EMD Serono, Genentech, Inc., Janssen Pharmaceutica Products, LP, Karyopharm Therapeutics Inc., and Kite Pharma, Inc.

Thomas M. Habermann, MD, Panel Member, has disclosed that he has no relevant financial relationships.

Stephen D. Smith, MD, Panel Member, has disclosed that he receives grant/research support from Acerta Pharma, AstraZeneca Pharmaceuticals LP, Denovo Biopharm, Genentech, Inc., Incyte Corporation, Merchk Sharp Dohme Corporation, and Portola Pharmaceuticals, Inc.; he is a consultant for AstraZeneca Pharmaceuticals LP; and his spouse receives grant/research support from Ayala Pharmaceuticals, Bristol-Myers Squibb Company, and Ignyta Inc.

Mary A. Dwyer, MS, CGC, Senior Manager, Guidelines, NCCN, has disclosed that she has no relevant financial relationships.

Hema Sundar, PhD, Oncology Scientist/Senior Medical Writer, NCCN, has disclosed that she has no relevant financial relationships.

To view all of the conflicts of interest for the panel, go to NCCN.org/disclosures/guidelinepanellisting.aspx.

This activity is supported by educational grants from AstraZeneca, Celgene Corporation, Clovis Oncology, Eisai, Genentech, Genomic Health, Inc., Novartis, Taiho Oncology, Inc., and TESARO. This activity is supported by an independent educational grant from AbbVie. This activity is supported by educational funding provided by Amgen. This activity is supported by an unrestricted educational grant from Gilead Sciences, Medical Affairs.

Overview

Diffuse large B-cell lymphomas (DLBCLs) and follicular lymphoma (FL) are the most common subtypes of B-cell non-Hodgkin’s lymphomas (NHLs) in adults, accounting for approximately 30% and 22%, respectively, of all newly diagnosed NHLs annually.1 Histologic transformation of FL to DLBCL (TFL) occurs in approximately 15% of patients, with an estimated annual rate of 2% to 3%, and is generally associated with a poor clinical outcome.24 In a pooled analysis of French and US cohorts of patients with newly diagnosed FL, TFL after diagnosis was the leading cause of death (77 of 140 deaths).5 Recent studies evaluating outcomes of patients with TFL in the rituximab era have reported improved survival in a subset of patients (eg, those with histologic transformation after early-stage FL, those with no previous exposure to chemotherapy or rituximab before histologic transformation).4,6,7

Recently, small molecule inhibitors targeting a variety of kinases involved in the B-cell receptor pathway have demonstrated promising activity for the treatment of relapsed/refractory FL. CAR T-cell therapy has emerged as a novel treatment option for relapsed/refractory DLBCL and TFL.

These NCCN Guidelines Insights highlight important updates to the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for B-Cell Lymphomas regarding the treatment of FL (relapsed/refractory disease and TFL) and DLBCL (relapsed/refractory disease).

Follicular Lymphoma

Anti–CD20 antibody–based chemoimmunotherapy is the standard initial treatment for newly diagnosed and relapsed/refractory FL. Frequently, patients with relapsed or progressive disease after first-line therapy will benefit from a second period of observation. Considerations and indications for treatment of relapsed/refractory or progressive disease include, among other factors, the modified GELF criteria, which include symptoms attributable to FL (not limited to B symptoms); threatened end-organ function; significant cytopenia secondary to lymphoma; bulky disease; splenomegaly; and steady progression over at least 6 months.

Progression of disease (POD) within ≤24 months of diagnosis and failure to achieve event-free survival at 12 months after initial treatment with chemoimmunotherapy have been identified as a prognostic indicators of poor survival.810 In the National LymphoCare Study, the 5-year overall survival (OS) rate was 50% for patients with POD ≤2 years after first-line therapy with R-CHOP (rituximab + cyclophosphamide/doxorubicin/vincristine/prednisone) compared with 90% for those without POD within ≤2 years.8 In a population-based analysis of relative survival of patients with FL compared with age- and sex-matched controls (from US and French datasets), patients who remained event-free 12 months after initial disease management had similar OS outcomes to the control group, whereas patients who experienced an event within 12 months had poor subsequent OS.9 Duration of response to first-line therapy is an important factor in the selection of second-line therapy. Options for treatment at first relapse include alternate non–cross-resistant chemoimmunotherapy regimens or combination lenalidomide + rituximab. Rituximab monotherapy may be appropriate for patients with late relapse as well, particularly if disease burden is low.

PI3K inhibitors (idelalisib, copanlisib, and duvelisib) have shown promising results in the treatment of relapsed/refractory disease after 2 prior therapies.1115 Idelalisib and copanlisib have also been shown to be effective in high-risk patients with disease relapse ≤2 years after first-line therapy.14,16

Based on the recent FDA approvals, idelalisib, copanlisib, and duvelisib are included as options for relapsed/refractory disease after 2 prior therapies. Evidence from clinical trials supporting this recommendation is discussed as follows.

Idelalisib

In a phase II multicenter single-arm study of 125 patients with rituximab- and alkylating agent–refractory indolent NHL (72 patients with FL), idelalisib resulted in tumor reductions in 90%, with an overall response rate (ORR) of 57% (6% complete response [CR]).11 Median duration of response and median progression-free survival (PFS) were 13 and 11 months, respectively. In a post hoc subgroup analysis of 72 patients with FL, the ORR was 56% (14% CR and 42% partial response [PR]) and the overall disease control rate was 87%.12 PFS rate at 12 months and estimated OS rate at 24 months were 43% and 70%, respectively. Grade ≥3 toxicities included neutropenia (27%), elevated alanine aminotransferase level (13%), diarrhea (13%), and pneumonia (7%), and thrombocytopenia (6%). In a post hoc analysis, median PFS was 8 months for patients with POD within 12 months compared with 14 months for those with POD between 12 and 24 months.16 The ORR, however, was not significantly different between the 2 groups (71% and 45%, respectively; P=.18).

Copanlisib

In a phase II study (CHRONOS-1) of 142 patients with relapsed/refractory indolent NHL after ≥2 prior lines of therapy, copanlisib resulted in an ORR of 59% in the subgroup of 104 patients with FL (14% CR and 44% PR) and median duration of response of 12 months. Median PFS and OS were 11 months and not reached, respectively.13 The 2-year follow-up data confirmed these results: ORR was 61% (20% CR and 39% PR) and median PFS and OS were was 13 and 43 months, respectively.14 Among the 98 patients with FL and POD within 24 months, the ORR was 60% (22% CR and 38% PR), which was not significantly different from the ORR of 59% for patients with POD after 24 months; however, the CR rate was higher in the group that experienced POD in <24 months (22%) compared with 18% in the group with POD after >24 months.14 Grade ≥3 toxicities included hyperglycemia (41%), neutropenia (24%), hypertension (24%), pneumonia (15%), thrombocytopenia (7%), and diarrhea (5%).

Duvelisib

In the phase II DYNAMO study evaluating the safety and efficacy of duvelisib in 129 patients with indolent NHL refractory to both rituximab and chemotherapy or radioimmunotherapy (RIT), duvelisib resulted in an ORR of 47% (42% for the subgroup with FL).15 Estimated median duration of response and median PFS were 10 and 9.5 months, respectively. Grade ≥3 toxicities included neutropenia (25%), anemia (15%), diarrhea (15%), and thrombocytopenia (12%).

Special Considerations for Use of PI3K Inhibitors

Diarrhea/colitis, hepatotoxicity, hyperglycemia, and transient cytopenias are the most common adverse events associated with PI3K inhibitors, and there are some differences in toxicities among the 3 available agents (Table 1).11,13,15 Recommendations for the monitoring and management of specific adverse events are outlined on pages 658 and 659 (NHODG-E).

Table 1.

Adverse Events of PI3K Inhibitors

Table 1.

Diffuse Large B-Cell Lymphoma

Second-line therapy followed by high-dose therapy with autologous stem cell rescue (HDT/ASCR) is appropriate treatment for relapsed/refractory DLBCL that is chemotherapy-sensitive at relapse in patients who are candidates for transplant.1720 Outcomes of relapsed/refractory DLBCL differ based on the response to initial therapy, timing of relapse, and opportunity to undergo HDT/ASCR. In a population-based study of 1,039 patients treated with anthracycline-based chemoimmunotherapy, 244 (23%) had relapsed disease that was subsequently re-treated.21 Across all therapies, the 4-year OS rate from relapse was 28%, and was 51% among those who underwent HDT/ASCR. The 4-year OS rate was 47% for patients with disease relapse after 12 months from initial diagnosis, whereas it was 13% for those with a transient or no response to initial therapy. The efficacy of second-line therapy is predicted by the second-line age-adjusted International Prognostic Index (IPI).22,23 Pretransplantation PET scans have been identified as predictive factors in patients undergoing HDT/ASCR, with positive PET results following second-line therapy predicting poor outcomes with transplantation.2426

Second-line combination chemotherapy is recommended for patients with an intention to proceed to transplant (see BCEL-7, page 656). Rituximab should be included if disease relapse occurs after a reasonable remission period (>6 months) and biopsy demonstrates continued CD20 expression; however; rituximab may be omitted in patients with primary refractory disease. Consolidation therapy with HDT/ASCR (category 1 for patients with CR) ± radiotherapy (RT) is recommended for patients with CR or PR to second-line therapy, if they are candidates for transplant.17,20 Involved-site RT (ISRT) before HDT/ASCR has been shown to result in good local disease control and improved outcome.27 Additional RT can be administered to limited sites with prior positive disease before or after HDT/ASCR. Allogeneic hematopoietic cell transplant (alloHCT) should be considered in select patients with mobilization failures and persistent bone marrow involvement or lack of adequate response to second-line therapy, although they should be in CR or near-CR at the time of transplant.

F3

Patients who are not candidates for transplant should ideally be treated in the context of a clinical trial. Standard options include gemcitabine-based regimens ± rituximab,28 bendamustine ± rituximab,29,30 or brentuximab vedotin for CD30-positive disease.31 Lenalidomide ± rituximab32,33 and ibrutinib34 are appropriate options at relapse, particularly for patients with non–germinal center B-cell–like (GCB) DLBCL.

Patients who are not candidates for transplant and experience a PR to second-line therapy, those with POD or refractory disease during second-line therapy (regardless of transplant eligibility), and those who experience disease relapse following HDT/ASCR or alloHCT should be managed with third-line systemic therapy, palliative ISRT, or best supportive care (see BCEL-8, page 657). Repeat biopsy should be strongly considered before additional therapy in patients who are noncandidates for transplant achieving PET-positive PR to second-line therapy, because PET positivity may represent posttreatment inflammation.

F4

However, patients experiencing POD after ≥2 prior lines of systemic therapy are unlikely to derive additional benefit from currently available systemic therapy options. The results of a recent multicenter, retrospective study (SCHOLAR-1) showed that outcomes are consistently poor among patients with refractory DLBCL.35 In this study evaluating the outcome of 636 patients with refractory DLBCL (defined as progressive or stable disease as best response at any point during chemotherapy [>4 cycles of first-line or 2 cycles of later-line therapy] or disease relapse at ≤12 months from HDT/ASCR), the pooled ORR was 26% (7% CR) to the next line of therapy, and median OS was 6 months.35 Response rates were much lower for patients with primary refractory disease or refractory disease after second-line or subsequent therapy (20% and 26%, respectively) compared with those with disease relapse ≤12 months from HDT/ASCR. Data from recent clinical trials (discussed in the following section) suggest that CAR T-cell therapy represents an improved treatment option for this group of patients.

CAR T-Cell Therapy

Axicabtagene ciloleucel and tisagenlecleucel are anti-CD19 CAR T-cell therapies FDA-approved for the treatment of adult patients with relapsed/refractory DLBCL, high-grade B-cell lymphomas (HGBL), and TFL (after ≥2 prior systemic therapy regimens).3638 Axicabtagene ciloleucel is also approved for relapsed/refractory primary mediastinal B-cell lymphoma (PMBL; after ≥2 prior systemic therapy regimens).

A multicenter phase II study (ZUMA-1) evaluated axicabtagene ciloleucel in patients with refractory DLBCL (n=81), TFL (n=30), or PMBL (n=8).36 Eligible patients had an absolute neutrophil count >1,000 cells/mcL, an absolute lymphocyte count >100 cells/mcL, a platelet count >75,000 cells/mcL, adequate organ function, no central nervous system (CNS) involvement, and no active infection. In the primary analysis that included 101 patients (78 with disease refractory to ≥2 lines of prior therapy and 21 with disease relapse after HDT/ASCR) who were evaluated 6 months after the infusion of axicabtagene ciloleucel, the ORR was 82% (54% CR and 28% PR). With a median follow-up of 15 months, the estimated PFS rate at 15 months and the OS rate at 18 months were 41% and 52%, respectively. The ORR was 81% (49% CR and 32% PR) and 83% (71% CR and 12% PR), respectively, for the subgroup of patients with DLBCL and TFL or PMBL. The 2-year follow-up data also confirmed these initial findings.37 After a median follow-up of 27 months, 39% of patients remained in ongoing remission and 51% remained alive. Median duration of response was 11 months and median PFS was 6 months.

The global multicenter phase II study (JULIET) evaluated tisagenlecleucel in patients with relapsed/refractory DLBCL (after ≥2 lines of therapy, including rituximab and an anthracycline) and TFL.38 Patients who were previously treated with anti-CD19 CAR T-cell therapy, received an alloHCT, had PMBL, or had active CNS involvement were excluded. In this study, 11 patients received an infusion with tisagenlecleucel. Among the 93 patients included in the efficacy analysis with ≥3 months of follow-up or had discontinued participation in the study before 3 months, the best ORR was 52% (40% CR and 12% PR). The estimated PFS rate at 12 months was 83% for patients who had a CR or PR at 3 months. The estimated OS rate at 12 months was 49% for all patients and 90% for those with a CR.

Axicabtagene ciloleucel and tisagenlecleucel result in responses across all subgroups, including double-hit/triple-hit lymphomas, GCB and non-GCB subtypes, and patients with either relapsed or primary refractory disease.3638 CD19 expression levels did not correlate with response. The NCCN Guidelines recommend CAR T-cell therapy (axicabtagene ciloleucel or tisagenlecleucel) for patients achieving PR after second-line therapy (regardless of their eligibility for transplant) and for those with disease relapse after achieving CR to second-line therapy or with progressive disease (see BCEL-7, opposite page, and BCEL-8, above).

Guidance for Treatment With CAR T-Cell Therapy

Axicabtagene ciloleucel and tisagenlecleucel should only be dispensed and administered in healthcare facilities that are enrolled in and comply with the Risk Evaluation and Mitigation Strategies (REMS) requirements.

Cytokine-release syndrome (CRS) and neurologic toxicity (also known as CAR T-cell–related encephalopathy syndrome [CRES]) are the most frequent life- threatening toxicities associated with CAR T-cell therapy. In the ZUMA-1 trial, CRS and neurologic toxicity of any grade were reported in 93% and 64% of patients, respectively.36,37 Median time from infusion to onset of symptoms was 2 and 5 days, respectively, with a median duration of 8 and 17 days, respectively. Grade ≥3 CRS and neurologic events occurred in 13% and 28% of the patients, respectively. Pyrexia (11%), hypoxia (9%), and hypotension (9%) were the most common symptoms of grade ≥3 CRS. Median time to onset of CRS was 2 days after infusion, and median time to resolution was 8 days. Encephalopathy (21%), confusion (9%), aphasia (7%), and somnolence (7%) were the most common grade ≥3 neurologic events. The incidence of grade ≥3 CRS and neurologic events decreased over the course of the study (occurring in 12% and 31% of patients, respectively, after a median follow-up of 15 months). In the JULIET trial, CRS and neurologic toxicity of any grade were reported in 58% and 21% of patients, respectively,38 and in 22% and 12%, respectively, for grade ≥3. Median time from infusion to onset of symptoms was 2 and 6 days, respectively, with a median duration of 7 and 14 days, respectively. Notably, different toxicity scales were used for grading CRS in the ZUMA-1 and JULIET trials (grade 3 CRS in JULIET was similar to grade 2 in ZUMA-1); therefore, although rates of any CRS can be compared, the rates of severe (grade 3/4) CRS specifically cannot be directly compared with accuracy.

F1
F2

The anti–interleukin-6 (IL-6) receptor monoclonal antibody tocilizumab is highly effective for the management of CRS, inducing rapid reversal of symptoms in most patients.39 Tocilizumab is approved for the treatment of CRS occurring after CAR T-cell therapy, and its use has not been shown to have any impact on the efficacy of CAR T-cell therapy in terms of response rates or duration of response. Corticosteroids are also an important adjunctive treatment of CRS, in concert with tocilizumab. Corticosteroids are preferred for the management of neurologic toxicity if not associated with CRS, whereas tocilizumab in combination with corticosteroids is recommended for management of CRS occurring in tandem with neurologic toxicity.39 Use of corticosteroids for the management of toxicities associated with CAR T-cell therapy did not impact the efficacy of CAR T-cell therapy, although it has been to shown to impair T-cell function. CRS and neurologic toxicity should be managed based on the toxicity grade as outlined in the CAR T-cell therapy–related toxicities section of the NCCN Guidelines for Management of Immunotherapy-Related Toxicities.

Histologically Transformed FL

Risk Factors

Advanced-stage FL at diagnosis, high-risk Follicular Lymphoma IPI and IPI scores at diagnosis, elevated lactate dehydrogenase level, and B symptoms at diagnosis have been reported to be risk factors for TFL.24,6,40,41

Retrospective analyses have suggested that early initiation of treatment at diagnosis relative to observation may decrease the risk of transformation in some studies.4,6,40 However, in the randomized phase III Intergroup trial that evaluated the role of immediate treatment with rituximab versus observation in patients with advanced-stage, asymptomatic, low-tumor-burden FL, and that also addressed whether early intensive rituximab therapy would change the risk of histologic transformation, no difference in time to transformation or incidence of histologic transformation was detected among the 3 groups after a median follow-up of almost 4 years.42

Treatment Options

No data from randomized studies support the optimal treatment of patients with TFL, because clinical trials have often excluded this group of patients. Results from retrospective cohort studies suggest that rituximab monotherapy or in combination with anthracycline-based chemotherapy is associated with improved OS.4,6,41 RIT has also been shown to induce high response rates and durable remissions in patients with TFL.43,44

In a series that reported the outcomes of 60 patients with biopsy-proven TFL, R-CHOP was the most commonly used treatment (n=35; 59%), associated with a median OS of 50 months.4 At a median follow-up of 60 months, the 5-year OS rate was 66% for patients with TFL treated with R-CHOP, which was similar to the outcome of patients with de novo DLBCL treated with R-CHOP in the validation cohort (5-year OS rate of 73%). In the National LymphoCare Study that included 147 pathologically confirmed and 232 clinically suspected cases of TFL, most patients were treated with rituximab-based therapy (26% of patients received rituximab monotherapy and 35% received chemotherapy with rituximab).6 At a median follow-up of 7 years, median PFS and OS were 12 and 60 months, respectively, for patients with biopsy-proven TFL. In another study (PRIMA) that evaluated the outcomes of patients with FL who experienced histologic transformation after response to first-line chemoimmunotherapy, most with TFL received chemoimmunotherapy regimens recommended for DLBCL, with a 5-year OS rate of 48% to 50%.41

HDT/ASCR as consolidation therapy has been evaluated only in retrospective studies,4552 with some series reporting a survival benefit for patients who proceeded to transplant.48,50,52 AlloHCT has been shown to benefit select patients with disease relapse after HDT/ASCR, but it is also associated with significant transplant-related mortality.48,49,51,53 However, it should be noted that the efficacy HDT/ASCR or alloHCT in patients with TFL has not been confirmed in prospective controlled studies.

Histologic Transformation After Minimal or No Prior Chemotherapy

Anthracycline-based chemoimmunotherapy (with regimens recommended for first-line therapy for DLBCL, unless contraindicated) ± ISRT is recommended for TFL without double-hit cytogenetics (see FOLL-6 and FOLL-7, pages 652 and 653).4,6 Histologic transformation to HGBL with translocations of MYC with either BCL2 or BCL6, also called double- or triple-hit lymphoma, occurs in approximately 24% of patients and has been associated with inferior survival compared with TFL without double-hit cytogenetics.54,55 TFL with double-hit cytogenetics should be managed with more aggressive chemoimmunotherapy regimens, as recommended for HGBL with translocations of MYC and BCL2 and/or BCL6.

F5
F6

Consolidation therapy with HDT/ASCR ± ISRT (if not previously given) or observation are included as treatment options for patients achieving CR or PR to initial treatment.48,50,52 If transformation is coexisting with extensive FL, rituximab maintenance should be considered for patients achieving CR. Additional systemic therapy ± ISRT to induce CR should be considered for patients achieving PR, if proceeding to transplant. Repeat biopsy should be strongly considered before additional therapy in patients with PET-positive PR, because PET positivity may represent posttreatment inflammation. AlloHCT may be considered in selected patients.

RIT with ibritumomab tiuxetan or ISRT (for localized residual disease and/or residual FDG-avid disease not previously irradiated) are included are alternative treatment options for patients achieving PR.43,44 RIT with ibritumomab tiuxetan or best supportive care are recommended for patients with nonresponsive or progressive disease.43,44 Based on the FDA approval, CAR T-cell therapy (axicabtagene ciloleucel or tisagenlecleucel) is included as an option (only after ≥2 prior chemoimmunotherapy regimens for indolent or transformed disease) for patients achieving PR or those with nonresponsive or progressive disease.3638

Histologic Transformation After Multiple Lines of Prior Therapies

Enrollment in an appropriate clinical trial is the preferred option. In the absence of a suitable clinical trial, treatment options include chemoimmunotherapy (with regimens recommended for second-line therapy for DLBCL) ± ISRT, RIT with ibritumomab tiuxetan, ISRT, or best supportive care (FOLL-8, page 654).41,43,44 Based on the FDA approval, CAR T-cell therapy (axicabtagene ciloleucel or tisagenlecleucel) is included as an option for patients who have received ≥2 prior chemoimmunotherapy regimens for transformed disease.3638

F7

Consolidation therapy with HDT/ASCR ± ISRT (if not previously given) or observation are included as treatment options for patients who achieve CR.48,50,52 AlloHCT may be considered in selected patients. For patients experiencing PR to initial therapy of TFL, treatment options include second-line regimens for DLBCL, alloHCT ± ISRT in selected patients, CAR T-cell therapy (axicabtagene ciloleucel or tisagenlecleucel for patients who have received ≥2 prior chemoimmunotherapy regimens for transformed disease) if not previously given, or ISRT for localized residual disease and/or residual FDG-avid disease not previously irradiated. However, it should be noted that data on the efficacy of transplant in patients who have received CAR T-cell therapy are not available. HDT/ASCR is not recommended after CAR T-cell therapy; alloHCT could be considered but remains investigational.

Patients with nonresponsive or progressive disease can be treated with any of the treatment options (not received previously) if they are candidates for additional therapy.

References

  • 1.

    Al-Hamadani M, Habermann TM, Cerhan JR, et al.. Non-Hodgkin lymphoma subtype distribution, geodemographic patterns, and survival in the US: a longitudinal analysis of the National Cancer Data Base from 1998 to 2011. Am J Hematol 2015;90:790795.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 2.

    Al-Tourah AJ, Gill KK, Chhanabhai M, et al.. Population-based analysis of incidence and outcome of transformed non-Hodgkin’s lymphoma. J Clin Oncol 2008;26:51655169.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Conconi A, Ponzio C, Lobetti-Bodoni C, et al.. Incidence, risk factors and outcome of histological transformation in follicular lymphoma. Br J Haematol 2012;157:188196.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Link BK, Maurer MJ, Nowakowski GS, et al.. Rates and outcomes of follicular lymphoma transformation in the immunochemotherapy era: a report from the University of Iowa/Mayo Clinic Specialized Program of Research Excellence Molecular Epidemiology Resource. J Clin Oncol 2013;31:32723278.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 5.

    Sarkozy C, Maurer MJ, Link BK, et al.. Cause of death in follicular lymphoma in the first decade of the rituximab era: a pooled analysis of french and US cohorts. J Clin Oncol 2019;37:144152.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 6.

    Wagner-Johnston ND, Link BK, Byrtek M, et al.. Outcomes of transformed follicular lymphoma in the modern era: a report from the National LymphoCare Study (NLCS). Blood 2015;126:851857.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 7.

    Gleeson M, Hawkes EA, Peckitt C, et al.. Outcomes for transformed follicular lymphoma in the rituximab era: the Royal Marsden experience 2003-2013. Leuk Lymphoma 2017;58:18051813.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 8.

    Casulo C, Byrtek M, Dawson KL, et al.. Early relapse of follicular lymphoma after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone defines patients at high risk for death: an analysis from the National LymphoCare Study. J Clin Oncol 2015;33:25162522.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Maurer MJ, Bachy E, Ghesquières H, et al.. Early event status informs subsequent outcome in newly diagnosed follicular lymphoma. Am J Hematol 2016;91:10961101.

  • 10.

    Casulo C, Le-Rademacher J, Dixon J, et al.. Validation of POD24 as a robust early clinical endpoint of poor survival in follicular lymphoma: results from the Follicular Lymphoma Analysis of Surrogacy Hypothesis (FLASH) investigation using individual data from 5,453 patients on 13 clinical trials [abstract]. Blood 2017;130:Abstract 412.

    • Search Google Scholar
    • Export Citation
  • 11.

    Gopal AK, Kahl BS, de Vos S, et al.. PI3Kδ inhibition by idelalisib in patients with relapsed indolent lymphoma. N Engl J Med 2014;370:10081018.

  • 12.

    Salles G, Schuster SJ, de Vos S, et al.. Efficacy and safety of idelalisib in patients with relapsed, rituximab- and alkylating agent-refractory follicular lymphoma: a subgroup analysis of a phase 2 study. Haematologica 2017;102:e156159.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Dreyling M, Santoro A, Mollica L, et al.. Phosphatidylinositol 3-kinase inhibition by copanlisib in relapsed or refractory indolent lymphoma. J Clin Oncol 2017;35:38983905.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Dreyling M, Santoro A, Mollica L, et al.. Long-term efficacy and safety from the copanlisib CHRONOS-1 study in patients with relapsed or refractory indolent B-cell lymphoma [abstract]. Blood 2018;132:Abstract 1595.

    • Search Google Scholar
    • Export Citation
  • 15.

    Flinn IW, Miller CB, Ardeshna KM, et al.. DYNAMO: a phase II study of duvelisib (IPI-145) in patients with refractory indolent non-Hodgkin lymphoma. J Clin Oncol 2019;37:912922.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Gopal AK, Kahl BS, Flowers CR, et al.. Idelalisib is effective in patients with high-risk follicular lymphoma and early relapse after initial chemoimmunotherapy. Blood 2017;129:30373039.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Philip T, Guglielmi C, Hagenbeek A, et al.. Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non-Hodgkin’s lymphoma. N Engl J Med 1995;333:15401545.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Vose JM, Zhang MJ, Rowlings PA, et al.. Autologous transplantation for diffuse aggressive non-Hodgkin’s lymphoma in patients never achieving remission: a report from the Autologous Blood and Marrow Transplant Registry. J Clin Oncol 2001;19:406413.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Rodriguez J, Caballero MD, Gutierrez A, et al.. Autologous stem-cell transplantation in diffuse large B-cell non-Hodgkin’s lymphoma not achieving complete response after induction chemotherapy: the GEL/TAMO experience. Ann Oncol 2004;15:15041509.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Mounier N, Canals C, Gisselbrecht C, et al.. High-dose therapy and autologous stem cell transplantation in first relapse for diffuse large B cell lymphoma in the rituximab era: an analysis based on data from the European Blood and Marrow Transplantation Registry. Biol Blood Marrow Transplant 2012;18:788793.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Farooq U, Maurer MJ, Thompson CA, et al.. Clinical heterogeneity of diffuse large B cell lymphoma following failure of front-line immunochemotherapy. Br J Haematol 2017;179:5060.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Hamlin PA, Zelenetz AD, Kewalramani T, et al.. Age-adjusted International Prognostic Index predicts autologous stem cell transplantation outcome for patients with relapsed or primary refractory diffuse large B-cell lymphoma. Blood 2003;102:19891996.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Lerner RE, Thomas W, Defor TE, et al.. The International Prognostic Index assessed at relapse predicts outcomes of autologous transplantation for diffuse large-cell non-Hodgkin’s lymphoma in second complete or partial remission. Biol Blood Marrow Transplant 2007;13:486492.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Spaepen K, Stroobants S, Dupont P, et al.. Prognostic value of pretransplantation positron emission tomography using fluorine 18-fluorodeoxyglucose in patients with aggressive lymphoma treated with high-dose chemotherapy and stem cell transplantation. Blood 2003;102:5359.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Derenzini E, Musuraca G, Fanti S, et al.. Pretransplantation positron emission tomography scan is the main predictor of autologous stem cell transplantation outcome in aggressive B-cell non-Hodgkin lymphoma. Cancer 2008;113:24962503.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Hoppe BS, Moskowitz CH, Zhang Z, et al.. The role of FDG-PET imaging and involved field radiotherapy in relapsed or refractory diffuse large B-cell lymphoma. Bone Marrow Transplant 2009;43:941948.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Hoppe BS, Moskowitz CH, Filippa DA, et al.. Involved-field radiotherapy before high-dose therapy and autologous stem-cell rescue in diffuse large-cell lymphoma: long-term disease control and toxicity. J Clin Oncol 2008;26:18581864.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Mounier N, El Gnaoui T, Tilly H, et al.. Rituximab plus gemcitabine and oxaliplatin in patients with refractory/relapsed diffuse large B-cell lymphoma who are not candidates for high-dose therapy. A phase II Lymphoma Study Association trial. Haematologica 2013;98:17261731.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Ohmachi K, Niitsu N, Uchida T, et al.. Multicenter phase II study of bendamustine plus rituximab in patients with relapsed or refractory diffuse large B-cell lymphoma. J Clin Oncol 2013;31:21032109.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Vacirca JL, Acs PI, Tabbara IA, et al.. Bendamustine combined with rituximab for patients with relapsed or refractory diffuse large B cell lymphoma. Ann Hematol 2014;93:403409.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Jacobsen ED, Sharman JP, Oki Y, et al.. Brentuximab vedotin demonstrates objective responses in a phase 2 study of relapsed/refractory DLBCL with variable CD30 expression. Blood 2015;125:13941402.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 32.

    Nowakowski GS, LaPlant B, Macon WR, et al.. Lenalidomide combined with R-CHOP overcomes negative prognostic impact of non-germinal center B-cell phenotype in newly diagnosed diffuse large B-Cell lymphoma: a phase II study. J Clin Oncol 2015;33:251257.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Czuczman MS, Trněný M, Davies A, et al.. A phase 2/3 multicenter, randomized, open-label study to compare the efficacy and safety of lenalidomide versus investigator’s choice in patients with relapsed or refractory diffuse large B-cell lymphoma. Clin Cancer Res 2017;23:41274137.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Wilson WH, Young RM, Schmitz R, et al.. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med 2015;21:922926.

  • 35.

    Crump M, Neelapu SS, Farooq U, et al.. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study. Blood 2017;130:18001808.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Neelapu SS, Locke FL, Bartlett NL, et al.. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017;377:25312544.

  • 37.

    Locke FL, Ghobadi A, Jacobson CA, et al.. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol 2019;20:3142.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Schuster SJ, Bishop MR, Tam CS, et al.. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2019;380:4556.

  • 39.

    Neelapu SS, Tummala S, Kebriaei P, et al.. Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat Rev Clin Oncol 2018;15:4762.

  • 40.

    Montoto S, Davies AJ, Matthews J, et al.. Risk and clinical implications of transformation of follicular lymphoma to diffuse large B-cell lymphoma. J Clin Oncol 2007;25:24262433.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41.

    Sarkozy C, Trneny M, Xerri L, et al.. Risk factors and outcomes for patients with follicular lymphoma who had histologic transformation after response to first-line immunochemotherapy in the PRIMA trial. J Clin Oncol 2016;34:25752582.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Ardeshna KM, Qian W, Smith P, et al.. Rituximab versus a watch-and-wait approach in patients with advanced-stage, asymptomatic, non-bulky follicular lymphoma: an open-label randomised phase 3 trial. Lancet Oncol 2014;15:424435.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43.

    Gordon LI, Witzig T, Molina A, et al.. Yttrium 90-labeled ibritumomab tiuxetan radioimmunotherapy produces high response rates and durable remissions in patients with previously treated B-cell lymphoma. Clin Lymphoma 2004;5:98101.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Mei M, Wondergem MJ, Palmer JM, et al.. Autologous transplantation for transformed non-Hodgkin lymphoma using an yttrium-90 ibritumomab tiuxetan conditioning regimen. Biol Blood Marrow Transplant 2014;20:20722075.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Smith SD, Bolwell BJ, Advani AS, et al.. High rate of survival in transformed lymphoma after autologous stem cell transplant: pathologic analysis and comparison with de novo diffuse large B-cell lymphoma. Leuk Lymphoma 2009;50:16251631.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Ban-Hoefen M, Kelly JL, Bernstein SH, et al.. High-dose therapy and autologous stem cell transplant for transformed non-Hodgkin lymphoma in the rituximab era. Leuk Lymphoma 2012;53:830835.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Reddy N, Oluwole O, Greer JP, et al.. Superior long-term outcome of patients with early transformation of non-Hodgkin lymphoma undergoing stem cell transplantation. Clin Lymphoma Myeloma Leuk 2012;12:406411.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48.

    Ban-Hoefen M, Vanderplas A, Crosby-Thompson AL, et al.. Transformed non-Hodgkin lymphoma in the rituximab era: analysis of the NCCN outcomes database. Br J Haematol 2013;163:487495.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Villa D, Crump M, Panzarella T, et al.. Autologous and allogeneic stem-cell transplantation for transformed follicular lymphoma: a report of the Canadian Blood and Marrow Transplant Group. J Clin Oncol 2013;31:11641171.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50.

    Villa D, Crump M, Keating A, et al.. Outcome of patients with transformed indolent non-Hodgkin lymphoma referred for autologous stem-cell transplantation. Ann Oncol 2013;24:16031609.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51.

    Wirk B, Fenske TS, Hamadani M, et al.. Outcomes of hematopoietic cell transplantation for diffuse large B cell lymphoma transformed from follicular lymphoma. Biol Blood Marrow Transplant 2014;20:951959.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Madsen C, Pedersen MB, Vase MO, et al.. Outcome determinants for transformed indolent lymphomas treated with or without autologous stem-cell transplantation. Ann Oncol 2015;26:393399.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53.

    Clavert A, Le Gouill S, Brissot E, et al.. Reduced-intensity conditioning allogeneic stem cell transplant for relapsed or transformed aggressive B-cell non-Hodgkin lymphoma. Leuk Lymphoma 2010;51:15021508.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54.

    Kridel R, Mottok A, Farinha P, et al.. Cell of origin of transformed follicular lymphoma. Blood 2015;126:21182127.

  • 55.

    McPhail ED, Maurer MJ, Macon WR, et al.. Inferior survival in high-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements is not associated with MYC/IG gene rearrangements. Haematologica 2018;103:18991907.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation

NCCN CATEGORIES OF EVIDENCE AND CONSENSUS

Category 1: Based upon high-level evidence, there is uniform NCCN consensus that the intervention is appropriate.

Category 2A: Based upon lower-level evidence, there is uniform NCCN consensus that the intervention is appropriate.

Category 2B: Based upon lower-level evidence, there is NCCN consensus that the intervention is appropriate.

Category 3: Based upon any level of evidence, there is major NCCN disagreement that the intervention is appropriate.

All recommendations are category 2A unless otherwise noted.

Clinical trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.

PLEASE NOTE

The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) are a statement of evidence and consensus of the authors regarding their views of currently accepted approaches to treatment. The NCCN Guidelines Insights highlight important changes in the NCCN Guidelines recommendations from previous versions. Colored markings in the algorithm show changes and the discussion aims to further the understanding of these changes by summarizing salient portions of the panel's discussion, including the literature reviewed.

The NCCN Guidelines Insights do not represent the full NCCN Guidelines; further, the National Comprehensive Cancer Network® (NCCN®) makes no representations or warranties of any kind regarding their content, use, or application of the NCCN Guidelines and NCCN Guidelines Insights and disclaims any responsibility for their application or use in any way.

The complete and most recent version of these guidelines is available free of charge at NCCN.org.

© National Comprehensive Cancer Network, Inc. 2019. All rights reserved. The NCCN Guidelines and the illustrations herein may not be reproduced in any form without the express written permission of NCCN.

  • Collapse
  • Expand
  • 1.

    Al-Hamadani M, Habermann TM, Cerhan JR, et al.. Non-Hodgkin lymphoma subtype distribution, geodemographic patterns, and survival in the US: a longitudinal analysis of the National Cancer Data Base from 1998 to 2011. Am J Hematol 2015;90:790795.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 2.

    Al-Tourah AJ, Gill KK, Chhanabhai M, et al.. Population-based analysis of incidence and outcome of transformed non-Hodgkin’s lymphoma. J Clin Oncol 2008;26:51655169.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Conconi A, Ponzio C, Lobetti-Bodoni C, et al.. Incidence, risk factors and outcome of histological transformation in follicular lymphoma. Br J Haematol 2012;157:188196.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Link BK, Maurer MJ, Nowakowski GS, et al.. Rates and outcomes of follicular lymphoma transformation in the immunochemotherapy era: a report from the University of Iowa/Mayo Clinic Specialized Program of Research Excellence Molecular Epidemiology Resource. J Clin Oncol 2013;31:32723278.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 5.

    Sarkozy C, Maurer MJ, Link BK, et al.. Cause of death in follicular lymphoma in the first decade of the rituximab era: a pooled analysis of french and US cohorts. J Clin Oncol 2019;37:144152.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 6.

    Wagner-Johnston ND, Link BK, Byrtek M, et al.. Outcomes of transformed follicular lymphoma in the modern era: a report from the National LymphoCare Study (NLCS). Blood 2015;126:851857.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 7.

    Gleeson M, Hawkes EA, Peckitt C, et al.. Outcomes for transformed follicular lymphoma in the rituximab era: the Royal Marsden experience 2003-2013. Leuk Lymphoma 2017;58:18051813.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 8.

    Casulo C, Byrtek M, Dawson KL, et al.. Early relapse of follicular lymphoma after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone defines patients at high risk for death: an analysis from the National LymphoCare Study. J Clin Oncol 2015;33:25162522.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Maurer MJ, Bachy E, Ghesquières H, et al.. Early event status informs subsequent outcome in newly diagnosed follicular lymphoma. Am J Hematol 2016;91:10961101.

  • 10.

    Casulo C, Le-Rademacher J, Dixon J, et al.. Validation of POD24 as a robust early clinical endpoint of poor survival in follicular lymphoma: results from the Follicular Lymphoma Analysis of Surrogacy Hypothesis (FLASH) investigation using individual data from 5,453 patients on 13 clinical trials [abstract]. Blood 2017;130:Abstract 412.

    • Search Google Scholar
    • Export Citation
  • 11.

    Gopal AK, Kahl BS, de Vos S, et al.. PI3Kδ inhibition by idelalisib in patients with relapsed indolent lymphoma. N Engl J Med 2014;370:10081018.

  • 12.

    Salles G, Schuster SJ, de Vos S, et al.. Efficacy and safety of idelalisib in patients with relapsed, rituximab- and alkylating agent-refractory follicular lymphoma: a subgroup analysis of a phase 2 study. Haematologica 2017;102:e156159.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 13.

    Dreyling M, Santoro A, Mollica L, et al.. Phosphatidylinositol 3-kinase inhibition by copanlisib in relapsed or refractory indolent lymphoma. J Clin Oncol 2017;35:38983905.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Dreyling M, Santoro A, Mollica L, et al.. Long-term efficacy and safety from the copanlisib CHRONOS-1 study in patients with relapsed or refractory indolent B-cell lymphoma [abstract]. Blood 2018;132:Abstract 1595.

    • Search Google Scholar
    • Export Citation
  • 15.

    Flinn IW, Miller CB, Ardeshna KM, et al.. DYNAMO: a phase II study of duvelisib (IPI-145) in patients with refractory indolent non-Hodgkin lymphoma. J Clin Oncol 2019;37:912922.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Gopal AK, Kahl BS, Flowers CR, et al.. Idelalisib is effective in patients with high-risk follicular lymphoma and early relapse after initial chemoimmunotherapy. Blood 2017;129:30373039.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Philip T, Guglielmi C, Hagenbeek A, et al.. Autologous bone marrow transplantation as compared with salvage chemotherapy in relapses of chemotherapy-sensitive non-Hodgkin’s lymphoma. N Engl J Med 1995;333:15401545.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Vose JM, Zhang MJ, Rowlings PA, et al.. Autologous transplantation for diffuse aggressive non-Hodgkin’s lymphoma in patients never achieving remission: a report from the Autologous Blood and Marrow Transplant Registry. J Clin Oncol 2001;19:406413.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Rodriguez J, Caballero MD, Gutierrez A, et al.. Autologous stem-cell transplantation in diffuse large B-cell non-Hodgkin’s lymphoma not achieving complete response after induction chemotherapy: the GEL/TAMO experience. Ann Oncol 2004;15:15041509.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Mounier N, Canals C, Gisselbrecht C, et al.. High-dose therapy and autologous stem cell transplantation in first relapse for diffuse large B cell lymphoma in the rituximab era: an analysis based on data from the European Blood and Marrow Transplantation Registry. Biol Blood Marrow Transplant 2012;18:788793.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Farooq U, Maurer MJ, Thompson CA, et al.. Clinical heterogeneity of diffuse large B cell lymphoma following failure of front-line immunochemotherapy. Br J Haematol 2017;179:5060.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Hamlin PA, Zelenetz AD, Kewalramani T, et al.. Age-adjusted International Prognostic Index predicts autologous stem cell transplantation outcome for patients with relapsed or primary refractory diffuse large B-cell lymphoma. Blood 2003;102:19891996.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Lerner RE, Thomas W, Defor TE, et al.. The International Prognostic Index assessed at relapse predicts outcomes of autologous transplantation for diffuse large-cell non-Hodgkin’s lymphoma in second complete or partial remission. Biol Blood Marrow Transplant 2007;13:486492.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Spaepen K, Stroobants S, Dupont P, et al.. Prognostic value of pretransplantation positron emission tomography using fluorine 18-fluorodeoxyglucose in patients with aggressive lymphoma treated with high-dose chemotherapy and stem cell transplantation. Blood 2003;102:5359.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Derenzini E, Musuraca G, Fanti S, et al.. Pretransplantation positron emission tomography scan is the main predictor of autologous stem cell transplantation outcome in aggressive B-cell non-Hodgkin lymphoma. Cancer 2008;113:24962503.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Hoppe BS, Moskowitz CH, Zhang Z, et al.. The role of FDG-PET imaging and involved field radiotherapy in relapsed or refractory diffuse large B-cell lymphoma. Bone Marrow Transplant 2009;43:941948.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Hoppe BS, Moskowitz CH, Filippa DA, et al.. Involved-field radiotherapy before high-dose therapy and autologous stem-cell rescue in diffuse large-cell lymphoma: long-term disease control and toxicity. J Clin Oncol 2008;26:18581864.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Mounier N, El Gnaoui T, Tilly H, et al.. Rituximab plus gemcitabine and oxaliplatin in patients with refractory/relapsed diffuse large B-cell lymphoma who are not candidates for high-dose therapy. A phase II Lymphoma Study Association trial. Haematologica 2013;98:17261731.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Ohmachi K, Niitsu N, Uchida T, et al.. Multicenter phase II study of bendamustine plus rituximab in patients with relapsed or refractory diffuse large B-cell lymphoma. J Clin Oncol 2013;31:21032109.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Vacirca JL, Acs PI, Tabbara IA, et al.. Bendamustine combined with rituximab for patients with relapsed or refractory diffuse large B cell lymphoma. Ann Hematol 2014;93:403409.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Jacobsen ED, Sharman JP, Oki Y, et al.. Brentuximab vedotin demonstrates objective responses in a phase 2 study of relapsed/refractory DLBCL with variable CD30 expression. Blood 2015;125:13941402.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 32.

    Nowakowski GS, LaPlant B, Macon WR, et al.. Lenalidomide combined with R-CHOP overcomes negative prognostic impact of non-germinal center B-cell phenotype in newly diagnosed diffuse large B-Cell lymphoma: a phase II study. J Clin Oncol 2015;33:251257.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Czuczman MS, Trněný M, Davies A, et al.. A phase 2/3 multicenter, randomized, open-label study to compare the efficacy and safety of lenalidomide versus investigator’s choice in patients with relapsed or refractory diffuse large B-cell lymphoma. Clin Cancer Res 2017;23:41274137.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Wilson WH, Young RM, Schmitz R, et al.. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med 2015;21:922926.

  • 35.

    Crump M, Neelapu SS, Farooq U, et al.. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study. Blood 2017;130:18001808.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Neelapu SS, Locke FL, Bartlett NL, et al.. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017;377:25312544.

  • 37.

    Locke FL, Ghobadi A, Jacobson CA, et al.. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol 2019;20:3142.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Schuster SJ, Bishop MR, Tam CS, et al.. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2019;380:4556.

  • 39.

    Neelapu SS, Tummala S, Kebriaei P, et al.. Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat Rev Clin Oncol 2018;15:4762.

  • 40.

    Montoto S, Davies AJ, Matthews J, et al.. Risk and clinical implications of transformation of follicular lymphoma to diffuse large B-cell lymphoma. J Clin Oncol 2007;25:24262433.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41.

    Sarkozy C, Trneny M, Xerri L, et al.. Risk factors and outcomes for patients with follicular lymphoma who had histologic transformation after response to first-line immunochemotherapy in the PRIMA trial. J Clin Oncol 2016;34:25752582.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Ardeshna KM, Qian W, Smith P, et al.. Rituximab versus a watch-and-wait approach in patients with advanced-stage, asymptomatic, non-bulky follicular lymphoma: an open-label randomised phase 3 trial. Lancet Oncol 2014;15:424435.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43.

    Gordon LI, Witzig T, Molina A, et al.. Yttrium 90-labeled ibritumomab tiuxetan radioimmunotherapy produces high response rates and durable remissions in patients with previously treated B-cell lymphoma. Clin Lymphoma 2004;5:98101.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Mei M, Wondergem MJ, Palmer JM, et al.. Autologous transplantation for transformed non-Hodgkin lymphoma using an yttrium-90 ibritumomab tiuxetan conditioning regimen. Biol Blood Marrow Transplant 2014;20:20722075.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Smith SD, Bolwell BJ, Advani AS, et al.. High rate of survival in transformed lymphoma after autologous stem cell transplant: pathologic analysis and comparison with de novo diffuse large B-cell lymphoma. Leuk Lymphoma 2009;50:16251631.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Ban-Hoefen M, Kelly JL, Bernstein SH, et al.. High-dose therapy and autologous stem cell transplant for transformed non-Hodgkin lymphoma in the rituximab era. Leuk Lymphoma 2012;53:830835.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Reddy N, Oluwole O, Greer JP, et al.. Superior long-term outcome of patients with early transformation of non-Hodgkin lymphoma undergoing stem cell transplantation. Clin Lymphoma Myeloma Leuk 2012;12:406411.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48.

    Ban-Hoefen M, Vanderplas A, Crosby-Thompson AL, et al.. Transformed non-Hodgkin lymphoma in the rituximab era: analysis of the NCCN outcomes database. Br J Haematol 2013;163:487495.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Villa D, Crump M, Panzarella T, et al.. Autologous and allogeneic stem-cell transplantation for transformed follicular lymphoma: a report of the Canadian Blood and Marrow Transplant Group. J Clin Oncol 2013;31:11641171.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50.

    Villa D, Crump M, Keating A, et al.. Outcome of patients with transformed indolent non-Hodgkin lymphoma referred for autologous stem-cell transplantation. Ann Oncol 2013;24:16031609.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51.

    Wirk B, Fenske TS, Hamadani M, et al.. Outcomes of hematopoietic cell transplantation for diffuse large B cell lymphoma transformed from follicular lymphoma. Biol Blood Marrow Transplant 2014;20:951959.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Madsen C, Pedersen MB, Vase MO, et al.. Outcome determinants for transformed indolent lymphomas treated with or without autologous stem-cell transplantation. Ann Oncol 2015;26:393399.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53.

    Clavert A, Le Gouill S, Brissot E, et al.. Reduced-intensity conditioning allogeneic stem cell transplant for relapsed or transformed aggressive B-cell non-Hodgkin lymphoma. Leuk Lymphoma 2010;51:15021508.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54.

    Kridel R, Mottok A, Farinha P, et al.. Cell of origin of transformed follicular lymphoma. Blood 2015;126:21182127.

  • 55.

    McPhail ED, Maurer MJ, Macon WR, et al.. Inferior survival in high-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements is not associated with MYC/IG gene rearrangements. Haematologica 2018;103:18991907.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 21256 3566 356
PDF Downloads 11593 2965 348
EPUB Downloads 0 0 0