Best Practice & Research Clinical Haematology 31 (2018) 49e56
Contents lists available at ScienceDirect
Best Practice & Research Clinical Haematology journal homepage: www.elsevier.com/locate/beha
Transformation of follicular lymphoma e Why does it happen and can it be prevented? Brian K. Link University of Iowa College of Medicine, Department of Internal Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
a r t i c l e i n f o
a b s t r a c t
Article history: Received 28 September 2017 Accepted 23 October 2017
Follicular lymphoma is a clinical disease with a multitude of presentations and behaviors. Although infrequent, transformation of follicular lymphoma to a more aggressive behaving subtype e prototypically diffuse large B-cell lymphoma e confers a substantially adverse prognosis. There is no consensus for optimal management after transformation is recognized. Historically considered a distinct clinical event, this review highlights the multiple subclinical transformational events that either variably or cumulatively result in clinical recognition of transformed follicular lymphoma. Known and suspected events include genetic and epigenetic perturbations, metabolomic changes, and alterations in the microenvironment. This diverse spectrum of pathways leads to heterogeneous clinical presentations and outcomes of transformed follicular lymphoma. Current options for prevention of transformation are limited to known strategies of managing follicular lymphoma before the transformation is recognized. Although most retrospectively analyzed studies suggest an association of lower transformation rates with early systemic therapy, specific components of therapy such as anti-CD20 antibodies, anthracyclines, or purine analogues are less strongly associated with “preventative’ value. Thus, the goal of preventing transformation is of limited value among all factors that go into decisions on early management of follicular lymphoma. Future opportunities to prevent clinical evidence of transformation will benefit from early detection of markers of subclinical transformation and development of therapies to specifically target the biology implied by those markers. © 2017 Published by Elsevier Ltd.
Keywords: Follicular lymphoma Transformed lymphoma Follicular transformation Review
1. Transformation of follicular lymphoma 1.1. Why is it of interest and what is the scope of the problem? Follicular lymphoma (FL) is a disease commonly characterized as incurable, though relatively slow in pace of growth with a median life expectancy of 15e20 years [1], and systemically treatable with expectations of somewhat extended response at the cost of limited toxicity [2]. As patients assimilate and adjust to the horror of incurable cancer, the lymphoma care team often prioritizes education to provide some comfort e especially since systemic therapy may not be indicated early after diagnosis. Somewhere in the educational effort, following “indolent”, “low grade” “responsive to therapy” “long remissions” “gentle treatments” and “many years”, the conversation gets around to “transformation” much like a depiction of rainbow-
E-mail address:
[email protected]. https://doi.org/10.1016/j.beha.2017.10.005 1521-6926/© 2017 Published by Elsevier Ltd.
50
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
colored fish, crabs, coral and sea turtles as occupants of the reef gets around to sharks. Along with early treatment failure following immunochemotherapy [3e6], transformation to high grade lymphoma e prototypically diffuse large B-cell lymphoma e represents the greatest fear for patients, their loved ones, and medical teams as the anticipated long journey with FL is commenced. Several recent reports of large cohorts of varying origin and methodology have provided a fairly consistent depiction of the frequency and implications of follicular lymphoma transformation (tFL) [6e17]. In the setting of immunotherapy and chemotherapy treatment options, the risk of transformation to DLBCL is 2e3% per year for FL patients for at least the first 10 years after which the data become less reliable though some series hint at lower rates farther out from diagnosis [14]. Outcomes following transformation are unfortunate and inferior to those of patient with FL who do not experience transformation but are no longer recognized as universally catastrophic as they had been in the twentieth century with median survival after transformation in most series between 4 and 5 years [7,9,13,15,16]. Survival is reported to be better for patients with lower FLIPI [18] scores, those previously anthracycline naïve, and for patients who transform later following FL diagnosis as opposed to transformation within the first 18 months [7,12,13]. Management strategies following recognition of transformation are numerous and without much consensus or consistent success [9,12,13,15,16]. Given the generally poor outcomes and unsatisfactory understanding of optimal management after transformation is clinically recognized, seeking better understanding of cause and potential prevention of clinically apparent transformation of follicular lymphoma is warranted. Two central tenets of this review will be heightened appreciation of the diversity of tFL and of the complex dynamics of preclinical transformation events. 2. Why does transformation happen? 2.1. Transformed follicular lymphoma is a heterogeneous disease with heterogeneous origins (Table 1) A simple definition for transformed follicular lymphoma is without universal consensus. The gold standard for diagnosing transformation in FL is generally recognized as biopsies demonstrating: a) FL grade 1, 2, or 3a and b) at a distant place or time histological demonstration of an increase in the proportion of large centroblasts infiltrating lymph nodes leading to effacement of the follicular architecture [19]. Strictly speaking these histologies should be confirmed to be clonally related. This gold standard is often adapted loosely in clinical series and in clinical practice. Such biopsies may be sequential, and some investigators historically restricted the definition of tFL to those scenarios where such biopsies were separated by at least 6 months [10,20,21]. Some newer published series of tFL, however, evaluated patients with concurrent identification of follicular lymphoma and diffuse large B-cell Lymphoma (DLBCL) either in a single biopsy [16] (alternatively referred to by others as composite lymphoma) [22,23] or in multiple synchronous biopsies (alternatively referred to as discordant lymphoma) [23,24]. Furthermore, some investigators accept a clinical definition of transformation in patients for whom no biopsy demonstrating DLBCL is available. Clinical criteria proposed by Al-Tourah et al. including a sudden rise in LDH, rapid discordant localized nodal growth, new B symptoms, hypercalcemia, or new extranodal sites of disease are the most often cited for inclusion in tFL series [8]. FDG-PET scans are also advocated by some as adjuvant determinants of transformation, with standardized uptake values between 10 and 17 proposed as a surrogate for pathological evidence of transformation [25e27]. Given recent advances in the appreciation of multiple pathways by which transformation can emerge from subclinical genetic alterations to the full blown clinical manifestations, this review will adapt an inclusive attitude when considering tFL and will endeavor to point out examples with a marginal or conditional definition for tFl when relevant. Histologic diversity of follicular lymphoma upon transformation is evident with the most common histology being DLBCL [8,19] followed by the entity previously categorized as Burkitt-like lymphoma, subsequently as B-cell lymphoma unclassifiable with features intermediate between DLBCL and Burkitt lymphoma, and most recently as high grade B cell lymphoma NOS [28e30]. Uncommonly, histologic and immunophenotypic characteristics of tFL overlap with acute lymphoblastic leukemia/lymphoma or other blastoid histologies [31,32]. Even the DLBCL histologies can be diverse with 80% marking as germinal b-cell like origin, yet 16% marking as activated b-cell like origin [33], and MYC rearrangement or overexpression is seen in nearly half of biopsies [34]. Clinical diversity of FL upon transformation is evident by timing of the recognition of transformation. Some authors recognize transformation at the time of lymphoma diagnosis when both DLBCL and FL histologies are found at initial evaluation. In the largest reported series of these “de novo transformed” patients, the National LymphoCare Study (NLCS) demonstrated probability of survival with de novo transformation was favorable when compared to patients with a more traditionally defined sequential transformation and not clearly different than FL patients in whom no transforming event was identified [16]. In another series, patients with recognized transformation within 18 months of FL diagnosis had a distinctly adverse 5 year survival rate relative to those with later transformation (66% vs 22%) [13]. Additionally, some patients are immunochemotherapy naïve at clinical transformation, some are simply chemotherapy naïve, and others have variable prior exposure to anthracyclines, alkylating agents or purine analogues [7,8,11,13,14,16,17]. Genetic diversity of tFL is profound with studies suggesting the transforming biology cannot be accounted for by disruptions of a unifying pathway. Early studies implicated acquisition of mutations in p53 [35,36] or p16 [37] in paired biopsy samples and at least altered gene expression if not frank mutation in MYC in many cases [38,39]. More recently, eloquent dissection of abnormalities in MLL2, EZH2, and CREBBP are suggestive of epigenetic (chromatin regulating) drivers of transformation in some patients [40e42], while alterations in MYC or CDKN2A/B in other patient samples implicate
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
51
Table 1 Diversity in transformed follicular lymphoma. Category
Variable
Frequency
reference
Histology
DLBCL Germinal b-cell like cell of origin Activated b-cell like cell of origin MYC rearranged or overexpressed High Grade B cell Lymphoma NOS Blastoid variant Recognition of FL and aggressive histology synchronous composite discordant sequential <18 months from FL diagnosis >18 months from FL diagnosis Therapy prior to transformation None Immunotherapy only Anthracycline naive Mutations in p53 Mutations in p16 Epigenetic drivers (MLL2, EZH2, and CREBBP) Cell cycle regulators (MYC, CDKN2A/B) Mitochondrial activity (peptide deformylase) CD4þ localization of cells within vs between follicles PD1þ T-cells
80% 80% 16% 50% 14% 1%
[8,19] [33] [33] [34] [19,28] [31,32]
10% NR NR 90% NR NR
[16]
Clinical
Genetic
Metabolomic Tumor Microenvironment
[13,16]
[9,12,13,16] 20e32% 20e30% 65e75% NR NR NR NR NR NR NR
[35,36] [37] [40e42] [41e43] [45e47] [62] [63]
regulators of proliferation and cell-cycle control [41e43]. Amplification of REL, BCL6, and BCL2 are other changes associated with transformation [44]. Metabolomic diversity is also hypothesized in tFL and other MYC edriven lymphomas, highlighted by investigations into the mitochondrial activity required to sustain tFL through the enzyme peptide deformylase (PDF) [42,45e47]. Though the pathway is under more careful study, it is included here because of the anti-lymphoma potential demonstrated by the PDF inhibitor actinonin [46,48]. The root of such diversity in tFL is likely found in the prevailing model of transformation. Follicular lymphoma is recognized to be a clinical disease emergent from a proliferation of clonal lymphocytes composed of innumerable subclones [49]. The subclonal genetic landscape indicates most subclones arise from distinct genetic events e usually from early FL “stem cells” e in a divergent rather than linear or sequential pattern, such that few perturbations are common to all subclones [50]. The preponderance of modern studies in tFL conclude that subclones dominant in biopsies demonstrating histological transformation acquired key genetic drivers of transformation early in the stem cell evolutionary process, and were genetically unique from the subclones dominant in the diagnostic FL histology [41,42,49,51]. Dynamic modeling demonstrated that the transformation-specific subclones rapidly attained clonal dominance in the transformed biopsy samples [50]. With multiple potential driver pathways and multiple early FL subclones on which to build, diversity of clinical, pathologic and genetic presentations is inevitable, without even accounting for variables in the host. Although articulate research has demonstrated convincingly that genetic alterations in the transformation of FL are phylogenetically early events, and at least in some cases already exist at the time of clinical FL diagnosis [50,52], there is no clear understanding of what causes those alterations or which of the alterations are necessary or sufficient to lead to clinically relevant transformed lymphoma behavior. Later discussion in this review will focus on the potential for prevention of tFL and for clinicians those opportunities will generally begin at a patient level once FL has been diagnosed. Thus, two key questions are how early can the markers suggestive of transformed biology be detected, and once detected; can clinical intervention impact emergence potential of the transformed subclone? In the absence of obvious target-specific therapy options, instinctive oncologist behavior might motivate the use of more aggressive cytotoxic chemotherapy combinations if subclones predictive of transformed behavior were identified prior to clinical transformed disease in an effort to “eradicate” the offending subclone. Alternatively, immunologist instincts might motivate minimal cytotoxic therapies in an effort to avoid disruption to any subclone control via the innate immune system or even to consider immune enhancing therapies including checkpoint inhibitors with direct or indirect T/NK cell augmentation [53,54]. Studies in emergence of secondary myeloid malignancies following chemotherapy suggest similar models of pre-existing subclones with malignant potential that emerge following cytotoxic chemotherapy [55,56]. Geneticist instincts might motivate any number of behaviors ranging from use of epigenetic modifiers such as histone de-acetylators [57] in an effort to prevent further evolution along the transformed pathway, to strict avoidance of therapies with further mutagenic potential. Although the role of host factors such as the tumor microenvironment are generally felt to be important in the biology of FL as assessed by gene expression [58], immunohistochemical methods relying on protein expression of tumor infiltrating immune cells have not yielded unifying results [59e61] and as a result the role of the tumor microenvironment in tFL is not as
52
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
well studied. Gene expression findings in FL patients developing tFL within 3 years or not were non-specifically suggestive of over-expression of immune-related genes while IHC findings included localization of CD4þ T cells within rather than between follicles more commonly in those patients with early tFL [62]. PD-1þ T cells have been associated with reduced risk of transformation [63]. A recent study from Norway evaluated paired biopsies from diagnosis and transformation in a cohort of 45 patients with tFL and compared findings with diagnostic biopsies from a matched cohort of patients with FL who's disease had not clinically transformed. In the tFL biopsies, PAX5þ tumor cells increased and CD3þT cells decreased compared to baseline samples. CD4þ, CD8þ, and CD57þ T cells also decreased. Higher total area scores of FOXP3þ T reg cells and intrafollicular CD4þ and CD57þ T cells at diagnosis were associated with higher risk of transformation and shorter time to transformation [64]. While in aggregate these findings lead credence to the likelihood that the immunologic milieu of the tumor microenvironment may play a role in development and emergence of tFL, improved methodology and more studies including larger numbers of patients with tFL will be needed to develop confidence-inspiring models. With a multitude of early sequential changes in the genetics, epigenetics, metabolomics, and the microenvironment implicated in the transformation of FL, it seems clear that current clinical and histological definitions of transformation along with diagnostic methodologies are inadequate for identifying the problem leading to undesirable outcomes for patients. A heightened recognition of the existence of diverse sub-clinical populations of lymphoma cells in various stages of transforming biology coupled with tools to detect those populations will be critical to development of strategies of more effective treatment or prevention. The accumulated genetic aberrancies associated with transformed biology make these subclinical populations good candidates for detection through assays of circulating tumor DNA (ctDNA) [65e67]. Plasma assays of ctDNA can provide information on total circulating clonal heterogeneity and assess molecular changes over time, whereas tissue biopsies are difficult to obtain serially and are subject to sample site specific biases. Assays of ctDNA utilizing rearranged VDJ sequences have already demonstrated utility in early detection of recurrent DLBCL [68,69]. Applications of ctDNA to monitoring follicular and other indolent lymphomas are under development [70].
3. Can transformation of follicular lymphoma be prevented? Many of the greatest successes in reduction of malignancy-related mortality in the previous century have been the result of prevention rather than successful therapies e cervical carcinoma and colon cancer with screening for precancerous lesions, lung cancer with avoidance of causative behavior and further anticipated reductions in cervical carcinoma, head and neck cancers and hepatoma with ongoing oncogenic viral management strategies [71e73]. Notably, these successes are seen in malignancies relatively resistant to systemic therapies as is tFL, although unlike tFL these successes are seen in malignancies with a high incidence. Given the generally poor outcomes of tFL and the unsatisfactory understanding of optimal management once clinically apparent, a strategy of prevention is appealing. Several series published to date provide the opportunity to examine if therapies currently employed give clues regarding a preventative strategy (Table 2). Very few of the series are prospective clinical trials, with the majority being retrospectively analyzed cohorts after either prospective or retrospective assembly, making uncontrolled or unmeasured variables hard to account for though many employ statistical efforts to aid as much as possible. The first obvious question is whether systemic therapy at time of FL diagnosis “prevents” or is at least associated with reduced risk of subsequent clinically evident transformation. This question is relevant because to initiate systemic therapy or not is often a fundamental decision addressed by patients and their providers with the decision based upon many factors. Two large prospectively assembled and retrospectively analyzed observational studies totalling over 3200 patients with Fl newly diagnosed after the availability of anti-CD20 antibody therapy both observed statistically reduced rates of tFL over time in
Table 2 Clinical strategies associated with risk of transformation (prevention). Therapy
Cohort summary
Associated effect on transformation rates
Initial therapy versus observation
>3500 FL pts prospectively collected and retrospectively analyzed 281 FL pts Retrospective series 463 pts prospectively randomized to rituximab versus observation >4000 FL pts retrospectively analyzed 1018 FL patients prospectively followed after choice of induction 286 FL pts retrospectively analyzed 463 pts prospectively randomized
Initial therapy favors protection from transformation [13,14,16]
Anthracycline as initial therapy Rituximab vs observation Rituximab maintenance
Observation favors protection from transformation Transformation events 11% observed versus 7% treated (p ¼ 0.19) after 4 years followup No protective effect seen with anthracycline use No protective effect seen with anthracycline use
Rituximab associated with lower transformation risk Transformation events 11% observed versus 7% treated (p ¼ 0.19) >3500 FL pts prospectively collected and retrospectively Maintenance rituximab associated with lower risk of analyzed transformation 1018 FL patients randomly assigned to maintenance rituximab Rituximab maintenance arm had fewer vs. observation after immunochemotherapy transformations (16 versus 24) 300 low burden FL patients assigned to rituximab maintenance 6 transformations in maintenance arm and 8 in vs retreatment at progression retreatment arm after 4.5 years followup
Reference
[17] [77] [7,11,13,16]
[13] [77] [13,16] [15] [78]
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
53
patients who had received initial therapy compared to those who deferred initial systemic therapy, with hazard ratios of approximately 0.6 after adjusting for other known risk factors [13,16]. This is consistent with a previous report of higher transformation rates following expectant observation in 325 FL patients diagnosed in the 20th century [14]. Two other observational series looking at the question demonstrated only non-significant trends toward protection with systemic therapy [7,8]. There are notably few prospective randomized trials of systemic therapy versus observation in newly diagnosed FL [74e76] and most don't report on subsequent tFL as an outcome, though a recent study from UK prospectively randomizing 463 patients with low risk FL to observation versus rituximab monotherapy reported 33 clinically apparent transformation events with a non-significant trend toward protection in the treated population (11% obs vs. 7% treated p ¼ 0.19) after just 4 years median followup [77]. In total, the preponderance of accumulated published data, though not conclusive, support the notion that early therapy reduces, though certainly doesn't effectively prevent risk of subsequent clinically apparent transformation. The next logical question is amongst the systemic therapies available, is there a drug or class of drugs, or maybe even a measure of therapy intensity, that is more effective than the others in preventing clinically apparent tFL. Two commonly studied therapy subsets in the above reports are the anthracyclines and anti CD20 antibodies. Anthracycline use as initial therapy had inconsistent association with transformation in the two larger and several smaller assembled cohorts. Authors from the University of Iowa/Mayo Clinic series and other series described no correlation between anthracycline treatment and rate of transformation, while the NLCS group showed lower rates of transformation when any anthracycline-containing regimen was compared to regimens void of anthracycline, but not when R-CHOP was compared specifically to R-COP [7,11,13,16]. Just one study described lower rates of subsequent clinically apparent tFL when anthracycline-based therapy was initiated compared to purine-based therapy [8]. The best opportunity to evaluate the role of early anthracycline use in prevention of subsequent clinically apparent transformation comes from the PRIMA (Rituximab Maintenance for 2 Years in Patients With High Tumor Burden Follicular Lymphoma Responding to Rituximab Plus Chemotherapy) trial. This prospective clinical trial with predetermined eligibility criteria and enrollment procedures ensures a relatively homogenous patient population with standardized baseline clinical assessments to minimize missing data. Initial management is relatively consistent (defined in this case as immunochemotherapy) allowing for comparison within the subtly different immunochemotherapy choices, such as anthracycline or not. The randomized variable of maintenance rituximab versus observation after response offers a unique opportunity to explore the role of this management option on incidence or outcome of subsequent transformation. With 40 histologically documented transformations among over 1000 FL patients initially treated with a variety of non-randomized immunochemotherapy regimens, no association was seen between use of anthracycline and subsequent transformation [15]. Several larger studies make at least an indirect argument for rituximab influencing transformation rates. In the Iowa/Mayo report initial therapy with rituximab monotherapy was associated with lower rates of transformation than initial observation, while both the NLCS report and PRIMA analysis e again with the strongest control on variables e suggest lower rates of transformation when rituximab maintenance is employed after initial remission induction [13,15,16]. As described earlier, the only prospective randomized study of rituximab as monotherapy versus observation in newly diagnosed FL showed only a non-significant trend toward protection in the treated population after a brief 4-year median followup [77]. In the US cooperative group RESORT (Rituximab Extended Schedule Or Re-Treatment) randomized trial in low burden FL, 300 subjects responding to induction rituximab were randomized 1:1 to receive maintenance rituximab until relapse or rituximab upon retreatment for relapse only. Patients assigned to maintenance received substantially more rituximab, but again with only 14 transformation (6 in maintenance arm and 8 in retreatment arm) events after a median followup of 4.5 years, no conclusions can be made [78]. Among 111 previously treated FL patients who received radioimmunotherapy in the form of 90Yittrium ibritumomab tiuxetan, a remarkable fraction e 29% - experienced histological transformation with a median follow-up of just under 5 years. Notably, 12 of 25 patients that had received prior therapy with the purine analogue fludarabine experienced transformation [79]. Acknowledging the hazards of drawing conclusions from a single small study composed of multiply treated FL patients (a population who's transformation incidence is not well characterized), this report coupled with the previously mentioned series in which purine analogues were associated with higher transformation events than anthracyclines at least somewhat weakens enthusiasm for considering either radioimmunotherapy or purine analogues as protective.
4. Summary on clinical prevention strategies How do we combine state of the art understanding of genetic and other possible pre-clinical events in the dynamic process of FL transformation with emerging potential therapies to prevent transformation of FL?. Option A is to nonspecifically suppress the FL stem cell pool to mathematically minimize the odds of a FL stem cell acquiring the necessary transformed biology. Option B is to identify very early pre-clinical transformed events e perhaps through monitoring of ctDNA and target the threatening subclone e ideally utilizing precision therapy if the biology of the subclone is understood well enough in the context of a broad array of potential therapies. Further down the road is the loftier goal of understanding FL stem cell biology and unlocking the biology of self-replication as is happening with the chronic myelogenous leukemia stem cell [80]. While waiting for scientists to unravel the puzzles needed to pursue option B, clinicians are currently limited to considering option A. Although probably effective in reducing transformation, the infrequency of transformation and outcomes following
54
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
therapy naïve transformations limit the motivation to invoke this strategy in all patients with FL, though it could be a deciding factor in patients who meet criteria for high risk. Conflict of interest In the past 2 years I have served on compensated DSMBs for AbbVie and Gilead, I am a consultant for Genentech and Celgene, and receive research support from Seattle Genetics. Funding This work was supported in part by grant no P50CA97274 from the National Institutes of Health, Bethesda MD, USA. Practice points
Transformed follicular lymphomas (FL) are genetically distinct from the diagnostic FL. Multiple genetic, epigenetic, and metabolomic factors drive transformation. Transformed FL presents with diverse clinical backgrounds and features. Systemic therapy early in FL is associated with mildy reduced transformation rates. No specific component of systemic therapy is recognized as clearly preventative.
References [1] Tan D, Horning SJ, Hoppe RT, Levy R, Rosenberg SA, Sigal BM, et al. Improvements in observed and relative survival in follicular grade 1-2 lymphoma during 4 decades: the Stanford University experience. Blood 2013;122(6):981e7. [2] Casulo C, Nastoupil L, Fowler NH, Friedberg JW, Flowers CR. Unmet needs in the first-line treatment of follicular lymphoma. Ann Oncol Off J Eur Soc Med Oncol 2017;28(9):2094e106. [3] Casulo C, Byrtek M, Dawson KL, Zhou X, Farber CM, Flowers CR, 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 lympho care study. J Clin Oncol Off J Am Soc Clin Oncol 2015;33(23):2516e22. [4] Maurer MJ, Bachy E, Ghesquieres H, Ansell SM, Nowakowski GS, Thompson CA, et al. Early event status informs subsequent outcome in newly diagnosed follicular lymphoma. Am J Hematol 2016;91(11):1096e101. [5] Provencio M, Royuela A, Torrente M, Pollan M, Gomez-Codina J, Sabin P, et al. Prognostic value of event-free survival at 12 and 24 months and longterm mortality for non-Hodgkin follicular lymphoma patients: a study report from the Spanish Lymphoma Oncology Group. Cancer 2017;23(19): 3709e16. [6] Mozessohn L, Cheung MC, Crump M, Buckstein R, Berinstein N, Imrie K, et al. Chemoimmunotherapy resistant follicular lymphoma: predictors of resistance, association with transformation and prognosis. Leukemia Lymphoma 2014;55(11):2502e7. [7] Alonso-Alvarez S, Magnano L, Alcoceba M, Andrade-Campos M, Espinosa-Lara N, Rodriguez G, et al. Risk of, and survival following, histological transformation in follicular lymphoma in the rituximab era. A retrospective multicentre study by the Spanish GELTAMO group. Br J Haematol 2017; 178(5):699e708. [8] Al-Tourah AJ, Gill KK, Chhanabhai M, Hoskins PJ, Klasa RJ, Savage KJ, et al. Population-based analysis of incidence and outcome of transformed nonHodgkin's lymphoma. J Clin Oncol Off J Am Soc Clin Oncol 2008;26(32):5165e9. [9] Ban-Hoefen M, Vanderplas A, Crosby-Thompson AL, Abel GA, Czuczman MS, Gordon LI, et al. Transformed non-Hodgkin lymphoma in the rituximab era: analysis of the NCCN outcomes database. Br J Haematol 2013;163(4):487e95. [10] Bastion Y, Sebban C, Berger F, Felman P, Salles G, Dumontet C, et al. Incidence, predictive factors, and outcome of lymphoma transformation in follicular lymphoma patients. J Clin Oncol Off J Am Soc Clin Oncol 1997;15(4):1587e94. [11] Gine E, Montoto S, Bosch F, Arenillas L, Mercadal S, Villamor N, et al. The Follicular Lymphoma International Prognostic Index (FLIPI) and the histological subtype are the most important factors to predict histological transformation in follicular lymphoma. Ann Oncol Off J Eur Soc Med Oncol 2006;17(10):1539e45. [12] Gleeson M, Hawkes EA, Peckitt C, Wotherspoon A, Attygalle A, Sharma B, et al. Outcomes for transformed follicular lymphoma in the rituximab era: the Royal Marsden experience 2003e2013. Leukemia Lymphoma 2017;58(8):1805e13. [13] Link BK, Maurer MJ, Nowakowski GS, Ansell SM, Macon WR, Syrbu SI, et al. Rates and outcomes of follicular lymphoma transformation in the immunochemotherapy era: a report from the university of Iowa/MayoClinic specialized program of research excellence molecular epidemiology resource. J Clin Oncol Off J Am Soc Clin Oncol 2013;31(26):3272e8. [14] Montoto S, Davies AJ, Matthews J, Calaminici M, Norton AJ, Amess J, et al. Risk and clinical implications of transformation of follicular lymphoma to diffuse large B-cell lymphoma. J Clin Oncol Off J Am Soc Clin Oncol 2007;25(17):2426e33. [15] Sarkozy C, Trneny M, Xerri L, Wickham N, Feugier P, Leppa S, 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 Off J Am Soc Clin Oncol 2016;34(22):2575e82. [16] Wagner-Johnston ND, Link BK, Byrtek M, Dawson KL, Hainsworth J, Flowers CR, et al. Outcomes of transformed follicular lymphoma in the modern era: a report from the National LymphoCare Study (NLCS). Blood 2015;126(7):851e7. [17] Conconi A, Ponzio C, Lobetti-Bodoni C, Motta M, Rancoita PM, Stathis A, et al. Incidence, risk factors and outcome of histological transformation in follicular lymphoma. Br J Haematol 2012;157(2):188e96. [18] Federico M, Bellei M, Marcheselli L, Luminari S, Lopez-Guillermo A, Vitolo U, et al. Follicular lymphoma international prognostic index 2: a new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. J Clin Oncol Off J Am Soc Clin Oncol 2009;27(27):4555e62. [19] Lossos IS, Gascoyne RD. Transformation of follicular lymphoma. Best Pract Res Clin Haematol 2011;24(2):147e63. [20] Hubbard SM, Chabner BA, DeVita Jr VT, Simon R, Berard CW, Jones RB, et al. Histologic progression in non-Hodgkin's lymphoma. Blood 1982;59(2): 258e64. [21] Oviatt DL, Cousar JB, Collins RD, Flexner JM, Stein RS. Malignant lymphomas of follicular center cell origin in humans. V. Incidence, clinical features, and prognostic implications of transformation of small cleaved cell nodular lymphoma. Cancer 1984;53(5):1109e14. [22] Kim H, Hendrickson R, Dorfman RF. Composite lymphoma. Cancer 1977;40(3):959e76.
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
55
[23] Kansara R, Connors JM, Savage KJ, Gerrie AS, Scott DW, Slack GW, et al. Maintenance rituximab following induction R-CHOP chemotherapy in patients with composite or discordant, indolent and aggressive, B-cell non-Hodgkin lymphomas. Haematologica 2016;101(10):e411e4. [24] Sehn LH, Scott DW, Chhanabhai M, Berry B, Ruskova A, Berkahn L, et al. Impact of concordant and discordant bone marrow involvement on outcome in diffuse large B-cell lymphoma treated with R-CHOP. J Clin Oncol Off J Am Soc Clin Oncol 2011;29(11):1452e7. [25] Karam M, Novak L, Cyriac J, Ali A, Nazeer T, Nugent F. Role of fluorine-18 fluoro-deoxyglucose positron emission tomography scan in the evaluation and follow-up of patients with low-grade lymphomas. Cancer 2006;107(1):175e83. [26] Noy A, Schoder H, Gonen M, Weissler M, Ertelt K, Cohler C, et al. The majority of transformed lymphomas have high standardized uptake values (SUVs) on positron emission tomography (PET) scanning similar to diffuse large B-cell lymphoma (DLBCL). Ann Oncol Off J Eur Soc Med Oncol 2009;20(3): 508e12. [27] Schoder H, Noy A, Gonen M, Weng L, Green D, Erdi YE, et al. Intensity of 18 fluorodeoxyglucose uptake in positron emission tomography distinguishes between indolent and aggressive non-Hodgkin's lymphoma. J Clin Oncol Off J Am Soc Clin Oncol 2005;23(21):4643e51. [28] Carbone A, Gloghini A, Aiello A, Testi A, Cabras A. B-cell lymphomas with features intermediate between distinct pathologic entities. From pathogenesis to pathology. Hum Pathol 2010;41(5):621e31. [29] de Jong D. Novel lymphoid neoplasmsethe borderland between diffuse large B-cell lymphoma and Burkitt's lymphoma. Haematologica 2009;94(7): 894e6. [30] Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127(20):2375e90. [31] De Jong D, Voetdijk BM, Beverstock GC, van Ommen GJ, Willemze R, Kluin PM. Activation of the c-myc oncogene in a precursor-B-cell blast crisis of follicular lymphoma, presenting as composite lymphoma. N. Engl J Med 1988;318(21):1373e8. [32] Natkunam Y, Warnke RA, Zehnder JL, Jones CD, Milatovich-Cherry A, Cornbleet PJ. Blastic/blastoid transformation of follicular lymphoma: immunohistologic and molecular analyses of five cases. Am J Surg Pathol 2000;24(4):525e34. [33] Kridel R, Mottok A, Farinha P, Ben-Neriah S, Ennishi D, Zheng Y, et al. Cell of origin of transformed follicular lymphoma. Blood 2015;126(18):2118e27. [34] Aukema SM, van Pel R, Nagel I, Bens S, Siebert R, Rosati S, et al. MYC expression and translocation analyses in low-grade and transformed Follicular Lymphoma. Histopathology July 21, 2017 [Epub ahead of print]. [35] Lo Coco F, Gaidano G, Louie DC, Offit K, Chaganti RS, Dalla-Favera R. p53 mutations are associated with histologic transformation of follicular lymphoma. Blood 1993;82(8):2289e95. [36] Sander CA, Yano T, Clark HM, Harris C, Longo DL, Jaffe ES, et al. p53 mutation is associated with progression in follicular lymphomas. Blood 1993;82(7): 1994e2004. [37] Elenitoba-Johnson KS, Gascoyne RD, Lim MS, Chhanabai M, Jaffe ES, Raffeld M. Homozygous deletions at chromosome 9p21 involving p16 and p15 are associated with histologic progression in follicle center lymphoma. Blood 1998;91(12):4677e85. [38] Elenitoba-Johnson KS, Jenson SD, Abbott RT, Palais RA, Bohling SD, Lin Z, et al. Involvement of multiple signaling pathways in follicular lymphoma transformation: p38-mitogen-activated protein kinase as a target for therapy. Proc Natl Acad Sci U. S. A 2003;100(12):7259e64. [39] Lossos IS, Alizadeh AA, Diehn M, Warnke R, Thorstenson Y, Oefner PJ, et al. Transformation of follicular lymphoma to diffuse large-cell lymphoma: alternative patterns with increased or decreased expression of c-myc and its regulated genes. Proc Natl Acad Sci U. S. A 2002;99(13):8886e91. [40] Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 2011;476(7360):298e303. [41] Okosun J, Bodor C, Wang J, Araf S, Yang CY, Pan C, et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet 2014;46(2):176e81. [42] Pasqualucci L, Khiabanian H, Fangazio M, Vasishtha M, Messina M, Holmes AB, et al. Genetics of follicular lymphoma transformation. Cell Rep 2014; 6(1):130e40. [43] Davies AJ, Rosenwald A, Wright G, Lee A, Last KW, Weisenburger DD, et al. Transformation of follicular lymphoma to diffuse large B-cell lymphoma proceeds by distinct oncogenic mechanisms. Br J Haematol 2007;136(2):286e93. [44] Martinez-Climent JA, Alizadeh AA, Segraves R, Blesa D, Rubio-Moscardo F, Albertson DG, et al. Transformation of follicular lymphoma to diffuse large cell lymphoma is associated with a heterogeneous set of DNA copy number and gene expression alterations. Blood 2003;101(8):3109e17. [45] Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol 2012;13(12):780e8. [46] Lee MD, She Y, Soskis MJ, Borella CP, Gardner JR, Hayes PA, et al. Human mitochondrial peptide deformylase, a new anticancer target of actinoninbased antibiotics. J Clin Investig 2004;114(8):1107e16. [47] Tucker EJ, Hershman SG, Kohrer C, Belcher-Timme CA, Patel J, Goldberger OA, et al. Mutations in MTFMT underlie a human disorder of formylation causing impaired mitochondrial translation. Cell Metab 2011;14(3):428e34. [48] Sheth A, Escobar-Alvarez S, Gardner J, Ran L, Heaney ML, Scheinberg DA. Inhibition of human mitochondrial peptide deformylase causes apoptosis in c-myc-overexpressing hematopoietic cancers. Cell Death Dis 2014;5:e1152. [49] Carlotti E, Wrench D, Rosignoli G, Marzec J, Sangaralingam A, Hazanov L, et al. High throughput sequencing analysis of the immunoglobulin heavy chain gene from flow-sorted B cell sub-populations define the dynamics of follicular lymphoma clonal evolution. PloS One 2015;10(9):e0134833. [50] Kridel R, Chan FC, Mottok A, Boyle M, Farinha P, Tan K, et al. Histological transformation and progression in follicular lymphoma: a clonal evolution study. PLoS Med 2016;13(12):e1002197. [51] Green MR, Gentles AJ, Nair RV, Irish JM, Kihira S, Liu CL, et al. Hierarchy in somatic mutations arising during genomic evolution and progression of follicular lymphoma. Blood 2013;121(9):1604e11. [52] Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AF, Esfahani MS, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med 2016;8(364):364ra155. [53] Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12(4):252e64. [54] Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol Off J Am Soc Clin Oncol 2015;33(17):1974e82. [55] Fabiani E, Falconi G, Fianchi L, Criscuolo M, Ottone T, Cicconi L, et al. Clonal evolution in therapy-related neoplasms. Oncotarget 2017;8(7):12031e40. [56] Sperling AS, Gibson CJ, Ebert BL. The genetics of myelodysplastic syndrome: from clonal haematopoiesis to secondary leukaemia. Nat Rev Cancer 2017; 17(1):5e19. [57] Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci 2017;18(7). [58] Dave SS, Wright G, Tan B, Rosenwald A, Gascoyne RD, Chan WC, et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N. Engl J Med 2004;351(21):2159e69. [59] Carreras J, Lopez-Guillermo A, Fox BC, Colomo L, Martinez A, Roncador G, et al. High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma. Blood 2006;108(9):2957e64. [60] Farinha P, Al-Tourah A, Gill K, Klasa R, Connors JM, Gascoyne RD. The architectural pattern of FOXP3-positive T cells in follicular lymphoma is an independent predictor of survival and histologic transformation. Blood 2010;115(2):289e95. [61] Lee AM, Clear AJ, Calaminici M, Davies AJ, Jordan S, MacDougall F, et al. Number of CD4þ cells and location of forkhead box protein P3-positive cells in diagnostic follicular lymphoma tissue microarrays correlates with outcome. J Clin Oncol Off J Am Soc Clin Oncol 2006;24(31):5052e9. [62] Glas AM, Knoops L, Delahaye L, Kersten MJ, Kibbelaar RE, Wessels LA, et al. Gene-expression and immunohistochemical study of specific T-cell subsets and accessory cell types in the transformation and prognosis of follicular lymphoma. J Clin Oncol Off J Am Soc Clin Oncol 2007;25(4):390e8. [63] Carreras J, Lopez-Guillermo A, Roncador G, Villamor N, Colomo L, Martinez A, et al. High numbers of tumor-infiltrating programmed cell death 1positive regulatory lymphocytes are associated with improved overall survival in follicular lymphoma. J Clin Oncol Off J Am Soc Clin Oncol 2009;27(9): 1470e6.
56
B.K. Link / Best Practice & Research Clinical Haematology 31 (2018) 49e56
[64] Blaker YN, Spetalen S, Brodtkorb M, Lingjaerde OC, Beiske K, Ostenstad B, et al. The tumour microenvironment influences survival and time to transformation in follicular lymphoma in the rituximab era. Br J Haematol 2016;175(1):102e14. [65] Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6(224):224ra24. [66] Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14(9):985e90. [67] Patel KM, Tsui DW. The translational potential of circulating tumour DNA in oncology. Clin Biochem 2015;48(15):957e61. [68] Kurtz DM, Green MR, Bratman SV, Scherer F, Liu CL, Kunder CA, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood 2015;125(24):3679e87. [69] Roschewski M, Dunleavy K, Pittaluga S, Moorhead M, Pepin F, Kong K, et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol 2015;16(5):541e9. [70] Roschewski M, Staudt LM, Wilson WH. Dynamic monitoring of circulating tumor DNA in non-Hodgkin lymphoma. Blood 2016;127(25):3127e32. [71] Finn OJ. Vaccines for cancer prevention: a practical and feasible approach to the cancer epidemic. Cancer Immunol Res 2014;2(8):708e13. [72] Jemal A, Ward EM, Johnson CJ, Cronin KA, Ma J, Ryerson B, et al. Annual report to the nation on the status of cancer, 1975e2014, featuring survival. J Natl Cancer Inst 2017;109(9). [73] Tsu V, Jeronimo J. Saving the World's women from cervical cancer. N. Engl J Med 2016;374(26):2509e11. [74] Ardeshna KM, Smith P, Norton A, Hancock BW, Hoskin PJ, MacLennan KA, et al. Long-term effect of a watch and wait policy versus immediate systemic treatment for asymptomatic advanced-stage non-Hodgkin lymphoma: a randomised controlled trial. Lancet 2003;362(9383):516e22. [75] Brice P, Bastion Y, Lepage E, Brousse N, Haioun C, Moreau P, et al. Comparison in low-tumor-burden follicular lymphomas between an initial notreatment policy, prednimustine, or interferon alfa: a randomized study from the Groupe d'Etude des Lymphomes Folliculaires. Groupe d'Etude des Lymphomes de l'Adulte. J Clin Oncol Off J Am Soc Clin Oncol 1997;15(3):1110e7. [76] Young RC, Longo DL, Glatstein E, Ihde DC, Jaffe ES, DeVita Jr VT. The treatment of indolent lymphomas: watchful waiting v aggressive combined modality treatment. Semin Hematol 1988;25(2 Suppl 2):11e6. [77] Ardeshna KM, Qian W, Smith P, Braganca N, Lowry L, Patrick 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(4):424e35. [78] Kahl BS, Hong F, Williams ME, Gascoyne RD, Wagner LI, Krauss JC, et al. Rituximab extended schedule or re-treatment trial for low-tumor burden follicular lymphoma: eastern cooperative oncology group protocol e4402. J Clin Oncol Off J Am Soc Clin Oncol 2014;32(28):3096e102. [79] Epperla N, Pham AQ, Burnette BL, Wiseman GA, Habermann TM, Macon WR, et al. Risk of histological transformation and therapy-related myelodysplasia/acute myeloid leukaemia in patients receiving radioimmunotherapy for follicular lymphoma. Br J Haematol 2017;178(3):427e33. [80] Li F, He B, Ma X, Yu S, Bhave RR, Lentz SR, et al. Prostaglandin E1 and its analog misoprostol inhibit human CML stem cell self-renewal via EP4 receptor activation and repression of AP-1. Cell Stem Cell 2017;21(3):359e73.