Unmet needs in histological transformation of follicular lymphoma: a clinical and biological review
Review Article

Unmet needs in histological transformation of follicular lymphoma: a clinical and biological review

Miguel Alcoceba1,2,3*, Sara Alonso-Álvarez2,4*, María García-Álvarez1*, Alejandro Martín1,2,3#, M. Dolores Caballero1,2,3#

1Department of Hematology, University Hospital of Salamanca (HUS/IBSAL), Salamanca, Spain; 2Grupo Español de Linfomas y Trasplantes de Médula Ósea (GELTAMO), Spain; 3CIBERONC, Spain; 4Department of Hematology, Central University Hospital of Asturias (HUCA), Oviedo, Spain

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

*These authors contributed equally to this work.

#These authors contributed equally for the senior authorship.

Correspondence to: M. Dolores Caballero, PhD, MD. Transplant and Clinic Unit, Department of Hematology, University Hospital of Salamanca (HUS/IBSAL),Paseo de San Vicente, 58-182,37007, Salamanca,Spain. Email: cabarri@usal.es.

Abstract: Histological transformation, with its profound diagnostic and therapeutic implications, is considered to be one of the most unfavorable events in the natural history of follicular lymphoma (FL). No optimal treatment of transformed follicular lymphoma (tFL) has been identified because of its rarity and heterogeneous presentation, and the lack of data from randomized clinical trials. Consequently, its treatment has been based on that used in de novo diffuse large B-cell lymphomas. However, patients with tFL present a different behavior, with a worse clinical course and poor response to treatment, although their prognosis seems to have improved significantly since the introduction of rituximab into therapeutic schemes. There is no consensus definition of tFL, which prevents the genuine identification of transformation when a biopsy is unavailable. There are no biomarkers at diagnosis that accurately identify patients who will transform. A better knowledge of the pathways leading to transformation would clearly help to identify patients at risk of transformation, and to distinguish them from patients who will relapse with FL and from those who will remain disease-free. This would also facilitate the identification of new drug targets, the design of clinical trials and the treatment of transformation when it arises. In the present review we provide a comprehensive overview of the current open questions in the field of transformation of FL. Large-scale international collaborative studies are required to address these gaps in our knowledge.

Keywords: Transformed follicular lymphoma (tFL); histological transformation


Received: 28 August 2017; Accepted: 21 November 2017; Published: 14 December 2017.

doi: 10.21037/aol.2017.11.03


Introduction

Non-Hodgkin lymphoma (NHL) is the most frequent hematological malignancy, with the seventh highest incidence of all cancers of 12.5 new cases per 100,000 inhabitants per year (1). Follicular lymphoma (FL) is the most common indolent NHL, with an estimated incidence of 20–30% of all lymphomas in western countries (1,2). The emergence of new drugs, especially for immunotherapy in induction and maintenance regimens, has improved median survival to up to 20 years (3-5). However, despite the high response rates and the slow course of the disease, FL is still considered to be an incurable disease characterized by a pattern of multiple relapses, decreasing duration of response, and gradual acquisition of resistance to different drugs (1,2). Up to 20% of patients progress within 24 months of treatment, and half of them die within 5 years (6,7), while those who remain in complete remission at this time have a similar overall survival (OS) as the general population (8). Moreover, an undefined percentage of FL with an estimated risk of 3% per year, either as a first or as a later event after one or several treatment lines, will transform into an aggressive lymphoma, usually diffuse large B-cell lymphoma (DLBCL). Outcomes of patients with transformed follicular lymphoma (tFL) have tended to be poor, but several studies carried out in the rituximab era suggest that survival may be more favorable than previously described (9-11). The prediction of histological transformation at diagnosis remains a challenge (12).

In this review, we focus on the biological mechanisms that underlie the process of histological transformation of FL to DLBCL, and the therapeutic strategies available for its treatment. Our review does not address other atypical forms suggestive of presentation of histological transformation, such as FLs that are already undergoing transformation at diagnosis (at the same or at different sites), or those that undergo a process of reverse transformation, from an aggressive lymphoma to a lower grade. This is not to deny their importance, however, and we urge that they be reviewed, since they might have different clinical behaviors.


Definition of FL transformation

The gold standard definition of FL transformation is based on the histologically confirmed progression of grade 1, 2, or 3A FL to a high-grade lymphoma (10), consisting of a predominance of large cells that eradicate the follicular architecture.

Since the tumor is disseminated, it should be borne in mind that, in addition to the transformation area, several tumor areas might have a concurrent FL component. Therefore, a single biopsy may not represent the entire tumor (10,13). There are several reasons why exclusive use of a histological criterion may make the detection of cases of transformation less likely: inaccessibility of the tumor that prevents a biopsy being performed, lack of available metabolic tests guiding the puncture, patients’ own limitations, or refusal to have their lesion surgically excised. Indeed, according to previous reports only half of the potential biopsies were performed (14).

Clinical criteria have been proposed when a biopsy is not feasible. However, there is no consensus about these, since they vary between studies (15-17). These criteria are arbitrary, based on clinical observations in patients with transformed lymphoma (15-17), but are also common clinical symptoms of FL progression (15-18). Overall, these include rapid discordant lymphadenopathy growth, a rapid increase in lactate dehydrogenase levels to 2 or 3 times normal values, the novel involvement of unusual extranodal sites, new B symptoms, and hypercalcemia (15-17). Although patients with a strong clinical suspicion of transformation might be clinically comparable to those with histologically confirmed transformation (16,17), there is not enough evidence to justify considering them on an equal footing. In fact, in some cases in which clinical suspicion would lead to the diagnosis of tFL, pathology reports have confirmed FL relapse (14).

In cases with clinical suspicion of tFL, the high standardized uptake value (SUV) of 18[F] fluorodeoxyglucose positron emission tomography and computerized tomography (PET/CT) is correlated with more aggressive histology, and therefore could help predict the transformation area (19-23). Previous studies have indicated that a maximum SUV value (SUVmax) of more than 17 could be enough to predict transformation (21,22). However, the scarcity of FL patients in the two studies may have led to a high SUVmax being overlooked in some FL cases, and in that sense, an SUVmax >17 at FL diagnosis or relapse has been reported (23). Moreover, and considering only the FL patients included in both studies, about one-third of tFL patients have an SUVmax <17, and up to 20% have an SUVmax of <10 (21,22). Thus, PET/CT may be helpful in guiding biopsies for the detection of histological transformation (21), although given the overlap of the SUVmax of FL and DLBCL, better imaging tools could contribute to a more reliable tFL diagnosis.


Incidence of transformation

It is well known that some patients with FL may eventually transform into an aggressive lymphoma, commonly DLBCL or, less frequently, Burkitt lymphoma or undifferentiated B-cell high-grade lymphoma (10,24). The study of the cell of origin (COO) shows that most of the tFLs (∼80%) are of the germinal B-cell subtype (GCB-DLBCL) (25,26). Such transformation has also been observed in other indolent B-cell lymphoproliferative disorders (B-LPDs), such as chronic lymphocytic leukemia, marginal zone lymphoma, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia, and nodular lymphocyte-predominant Hodgkin lymphoma (10,11,27-31).

The incidence of tFL is not well established, mainly due to the use of the previously mentioned different criteria for defining and classifying transformation, cohort size, and duration of follow-up of the studies, among others (9,11,14-17,32-36) (Table 1). In addition, in most studies, clonality testing to distinguish between actual transformation to de novo DLBCL has not been carried out, thus including ~5–10% of cases that are clonally unrelated (37,38). Taking all these points into consideration, the incidence of tFL has been estimated to be 15–30% at 10 years in the pre-rituximab and rituximab eras, with a consistent risk of ~3% of tFL per year, although a recent work suggests a reduction in the rituximab era (16,17,36,39). The report of a 70% incidence in an autopsy series suggests that tFL is a common terminal event in FL (40).

Table 1

Incidence of transformation (only series with >200 cases are reported)

References Criteria of tFL Rituximab era n tFL (n) Proven biopsy (%) Follow-up (year) Incidence of transformation
Bastion (15) HT + clinical No 220 52 65 9 22% at 5 years
Gine (14) HT + clinical No 276 30 83 6.5 15% at 10 years
Montoto (9) HT No 325 88 100 15 28% at 10 years
Al-Tourah (16) HT + clinical No 600 170 63 9.1 3% annually
Conconi (32) HT No 281 37 100 10 15% at 10 years
Bains (33) HT + clinical * 237 34 74 7.4 18.5% at 10 years
Link (17) HT + clinical Yes 631 60 85 5 10.7% at 5 years
Wagner-Johnston (34) HT + clinical Yes 2,652 379 39 6.8 14.3% at 6.8 years
Sarkozy (35) HT Yes 1,017 40 100 6.1 4.1% at 6 years
Alonso-Álvarez (11) HT Yes 1,734 106 100 6.2 5% at 5 years; 8% at 10 years
Federico (36) HT Yes 7,405 439 100 5.5% at 5 years; 7.1% at 10 years

*, Limited-stage follicular lymphoma. HT, histological transformation; tFL, transformed follicular lymphoma.

In the rituximab era, only two large studies have analyzed the incidence of tFL using strictly histological criteria. First, in the PRIMA trial, which enrolled patients achieving partial or complete response after first-line treatment, showed that the incidence of tFL was 4% at 6 years (35). When estimating the incidence of transformation, we must bear in mind that Kaplan-Meier curves consider only one possible event, and the censored cases are an estimated risk in the future. Therefore, patients who die before transformation should not be censored, making a competing risk model a better choice for analyzing cumulative incidence (41). In the recently published study of the Spanish GELTAMO group, we calculated the cumulative incidence of tFL in a large cohort series, considering death as a competing risk, estimated as 5% at 5 years, and 8% at 10 years (11).

Whether the risk of tFL is constant over time is open to question. Two series in the pre-rituximab era found a plateau at ∼17 years (9,32), while most of the other studies find no such plateau (11,14-17,33-35). However, the former studies had a median follow-up of 10–15 years, while those of the latter studies were relatively shorter (<10 years). A longer follow-up in a large series is required to answer this question as it applies in the rituximab era.

The recent ARISTOTLE project launched by the European Lymphoma Institute (ELI), integrates the retrospective data of ten collaborative national groups studying lymphoma, including those of the PRIMA trial, and the GELTAMO studies, and is likely to go some way to providing answers to these outstanding questions (36). Preliminary results, considering histological criteria and only first events, suggest an incidence of tFL of 7% at 10 years (36). A reduction in the incidence of transformation at 10 years was observed in patients receiving rituximab in the induction and maintenance regimen, compared with those receiving it only at induction. A longer follow-up of the series is required to determine the role of maintenance in delaying or decreasing the risk of transformation (36).


One of the major challenges in the treatment of FL is to identify predictors of histological transformation at diagnosis (12). Previous studies have shown that patients with high-risk clinical factors (high FL international prognostic index—FLIPI) have a greater risk of transformation (9,11,14,17,35). However, a considerable proportion of patients with low-risk clinical factors will transform, while, conversely, some patients with multiple high-risk factors will not transform (9,11,17,35,42).

The suboptimal response to first-line treatment has also been associated with a higher risk of transformation in the pre-rituximab and rituximab eras (11,15,43). However, it is well documented that transformation can occur in responding and non-responding cases, and even in untreated patients (11,15,35). The fact that shorter survival was observed in patients who transformed within 18 months of diagnosis (17), suggests that the acquisition of certain alterations drives a faster progression of the disease in some tFL cases. Recurrent gene alterations in the transformation include TP53, CDKN2A/B, and MYC, which are common in relapsed/refractory patients of different hematological disorders, including FL, and probably cause the aggressiveness of the disease (42,44). Although our knowledge of the molecular mechanisms involved in FL transformation has increased in recent years, the etiology and pathogenesis of the transformation are still unclear.

It remains to be clarified whether early treatment is associated with lower transformation risk compared with a “watch and wait” strategy as has been suggested in several papers (9,17,34). The phase 3 clinical trial conducted by Ardeshna et al. (45) which randomized patients to early rituximab therapy or observation, is best suited to answering this question. With a median follow-up of almost 4 years, no difference in the time to transformation, or incidence of histological transformation, was detected, although a longer follow-up will be required to achieve definitive conclusions.


Pathogenesis of tFL

The advent of next-generation sequencing (NGS) has enabled the mutational landscape of different hematological malignancies to be determined. Most studies of NHL have focused on FL and/or DLBCL (46-48), and do not include tFL or reflect it as a subgroup; therefore, few studies have specifically addressed tFL (49,50). Most of these studies are largely descriptive and compare the condition with other disorders, meaning that few studies have investigated the clinical significance of the majority of these gene mutations (51-56).

Two studies have used NGS and copy-number alteration (CNA) to show that the tFL genomic landscape shares alterations with FL and GCB-DLBCL (e.g., CREBB, EZH2, and MLL2 mutations); alterations exclusive to tFL have also been described (e.g., CDKN2A/B deletions, EBF1 and TNFAIP3 mutations) that might have diagnostic and potentially therapeutic implications (49,50). The major limitation of both studies was the absence of an association between genetic alteration and clinical behavior or the risk of transformation. Although these studies imply that there is clonal evolution in tFL, which is consistent with a model of divergent transformation from a common progenitor cell rather than a lineal model, the small number of cases (12 and 10, respectively) prevents conclusive results being drawn (49,50). Indeed, various studies suggest that the presence of TP53 alterations is a risk factor for transformation in FL (42,49,50).

In a very recent study comparing FLs that progress or relapse with those that transform (either as an initial or subsequent event), transformation was the result of drastic changes of subclones present at very low frequencies (<1%) at diagnosis compared with progression, where the clone responsible was observed at diagnosis (42). These authors analyzed a custom 94-gene panel in 118 FL-tFL pairs and FL patients without transformation ≥5 years after diagnosis, identifying frequent genes in each group. Since most of the FL patients without transformation (>75%) had a low-risk FLIPI, a thorough comparison with patients with matched high-risk factors (high-FLIPI) at diagnosis has not been carried out. Another recent study combining CNA and NGS from 35 tFLs, identified, several deregulated pathways, including JAK-STAT, NF-kB, and immune surveillance (57).

In recent years, genome-wide analyses have identified the 6p21.3 region as a susceptibility region for a range of NHLs, including FL (58-60). The human leukocyte antigen (HLA) system, which is located in this region, has a central role in the anti-tumor immune response and in the apoptosis of lymphoma cells (61), and could therefore have a role in disease control. Earlier studies have shown a relationship between different HLA polymorphisms and a higher susceptibility to developing FL or DLBCL, including a group of HLA polymorphisms that confer a worse prognosis in DLBCL (62-65). However, no studies have investigated the role of HLA polymorphisms in the risk of transformation, while single-nucleotide polymorphisms (SNPs) within the 6p21 region, such as rs6457327, have been associated with both a higher risk of transformation and lower survival from transformation (66,67).


Composition of the microenvironment

The cellular microenvironment is important for the control, development, and proliferation of tumors. Molecular studies have demonstrated its involvement in various hematological malignancies and the current possibility of using molecules that inhibit the inhibitory signals (e.g., anti PD-1, or CTLA-4) of the immune response which act on tumor-infiltrating cells, suggest that the microenvironment is able to terminate the tumor cells (68,69).

CD4+ and regulatory T-cells (Tregs) have not been uniformly associated with the increased or decreased risk of transformation, either in the pre-rituximab era, or in the current scenario with new drugs. Published studies yielded apparently inconsistent findings, probably due to the different combinations of chemotherapy used. Taking this into consideration, in a cohort of differently treated patients, a follicular pattern of FOXP3+ Tregs was associated with an increased risk of transformation (70). Furthermore, CD4-positive T-helper cells, and specifically T-helper 1 cells, have more frequently been found within the neoplastic follicles of FL patients who underwent early transformation to an aggressive histology than in those who did not (71).

Similarly, a high level of tumor- or lymphoma-associated macrophages (TAMs) has been associated with an adverse outcome in the pre-rituximab era (chemotherapy-treated FL patients), with respect to PFS and OS (72,73). However, this association might be conditioned by the type of chemotherapy received (fludarabine- or cyclophosphamide-based) (74). Moreover, the most recent studies have detected that associations between macrophages and outcome might have changed after rituximab, so that more TAMs would be correlated with longer survival rates (75,76). Moreover, macrophages cannot be considered individually, since an abundance of tumor-associated mast cells has been linked to an adverse prognosis, even in cases with a favorable level of TAMs (77). The role of macrophages in predicting FL transformation in the current scenario in which immunochemotherapy coexists with a wide spectrum of new drugs is still unknown.

Finally, it is noted that increased angiogenesis in FL tumor samples is associated with worse survival and a higher risk of transformation (78).

Therefore, there is very limited information about the role of the composition of the microenvironment in the risk of transformation under the current scenario. Newer technologies have made it possible to determine not only the number and spatial distribution of tumor-associated cells, but also their gene expression profiles, and their different functional status. This leads us to conclude that it is time to comprehensively analyze these characteristics in large series in conjunction with other biological and clinical factors.


Therapeutic strategies in tFL

No prospective studies have investigated patients with tFL in the rituximab era, so treatment is generally based on that used in de novo DLBCL. Even though the same therapeutic schemes are used, tFL patients have been considered to behave differently from de novo DLBCL, with a worse clinical course, poorer response to treatment, and shorter survival, usually less than two years in the pre-rituximab era (10). The introduction of the monoclonal antibody rituximab improved de novo DLBCL prognosis (79-81), and also seems to have improved the prognosis of tFL patients, as shown in Table 2 (82-102), extending the median survival to 4–5 years (10,17,84). In fact, recent studies have not found any significantly different outcome between tFL and de novo DLBCL (97,103).

Table 2

Studies on outcomes of chemotherapy, auto-SCT, and allo-SCT in tFL

References Rituximab era tFL (n) PFS OS TRM Comments
Time (year) % Time (year) % Time %
Chemotherapy
   Kaminski (82) No 14 Median 13.9 months 4 20 Prospective clinical trial—RT + Tositumomab
   Gleeson* (83) Yes 70 5 40 5 64.5 Excluded 17 cases treated without rituximab (n=8) and with auto-SCT as consolidation (n=9). R-CHOP (n=55), 37% 5-year PFS and 64.3% 5-year OS
   Ban-Hoefen* (84) Yes 50 N.D. 2 53 Prospective study
   Villa* (85) Yes 53 5 40 5 61 Only <65 years patients; 32% treatment-naïve at HT
   Madsen (86) Yes 18 5 6 5 36 Only <67 year patients
Auto-SCT
   Foran (87) No 19 5 52 5 50 100 days 5
   Williams (88) No 50 5 30 5 51 100 days 8
   Andreadis (89) No 22 Median 1.4 years Median 4.6 years 100 days 2
   Sabloff (90) No 23 5 25 5 56 1 year 0
   Hamadani (91) Yes 24 5 33 5 52 100 days 8
   Smith (92) Yes 25 §3 64 3 63 100 days 0
   Eide (93) No 30 5 32 5 47 100 days 0 Prospective clinical trial
   Ban-Hoefen (94) Yes 16 2 59 2 82 100 days 0 Rituximab naïve better PFS
   Reddy (95) Yes 44 5 45 5 62 2 years 4.6 ≥1 line from HT to auto-SCT
   Ban-Hoefen* (84) Yes 50 N.D. 2 83 Overall 2 Prospective
   Villa* (85) Yes 97 5 55 5 65 100 days 2
   Wirk (96) Yes 108 5 35 5 50 1 year 8
   Kuruvilla (97) Yes 46 4 45 4 65 N.D. Prospective clinical trial; 58% of tFL received R as maintenance
   Madsen (86) Yes 33 5 53 5 62 N.D. 3 low-grade NHL; auto-SCT better when R-naïve at HT
Allo-SCT
   Rezvani (98) Yes 16 3 21 3 18 3 years 42 Non-MAC, auto-SCT previous in 44%
   Hamadani (99) Yes 8 4 56 4 66 100 days 12.5 75% MAC
   Ramadan (100) Yes 25 3 32 3 48 1 year 33 MAC
   Thomson (101) Yes 18 4 61 4 60 1 year 29 RIC, auto-SCT previous in 56%
   Clavert (102) Yes 9 5 89 5 67 1 year 25 RIC; auto-SCT + allo-SCT; numbers estimated for the 9 tFL; 68% PFS and OS at 4 years for the whole series (n=19)
   Reddy (95) Yes 7 5 45 5 69 2 years 31.4 43% MAC
   Ban Hoefen* (84) Yes 18 N.D. 2 65 Overall 22 Prospective
   Villa* (85) Yes 22 5 46 5 46 100 days 5 >95% MAC
   Wirk (96) Yes 22; 11 3; 3 11; 48 3; 3 11; 67 1; 1 57; 0 22 MAC; 11 RIC

* includes tFL without prior treatment (21% in chemotherapy in Gleeson; 32% in chemotherapy, 10% in auto-SCT, and 11% in allo-SCT in Ban Hofen; 32%, 16%, and 14% respectively in Villa); includes all indolent transformed NHLs (percentage of tFL as follows: 86% in Ban Hofen; 72% + 20% tFL at diagnosis in Smith; 90% in Kuruvilla; 84% in Ramadan); , event-free survival; §, relapse-free survival; , referred to the total series, not restricted to tFL (n=40 including 15 tFL at diagnosis in Ramadan; n=48 including 30 relapsed DLBCL in Thomson). N.D., not described; MAC, myeloablative conditioning; OS, overall survival; PFS, progression-free survival; RIC, reduced intensity conditioning; RT, radiotherapy; tFL, transformed follicular lymphoma; TRM, transplant-related mortality.

It seems clear that the treatment should not be the same for patients who have or have not previously been treated for FL. Thus, a therapeutic model has been proposed that recommends treating treatment-naïve patients with rituximab plus chemotherapy (R-CHOP), as if they were de novo DLBCL cases (104). Consolidation with autologous stem cell transplantation (auto-SCT) has been a standard therapy for tFL patients, as it has been for relapsed chemosensitive NHL (105). However, given the good results obtained with R-CHOP in chemotherapy-naïve patients, the need for auto-SCT has been called into question, and it could be obviated in patients who achieve complete remission after R-CHOP (83,103).

A different scenario arises in relation to patients with tFL who have already received R-CHOP during their FL stage. In these cases, initial therapy should be different from R-CHOP, using platinum-based regimens, and autotransplant intensification may be appropriate (84-97).

The role of allogeneic stem-cell transplantation (allo-SCT) in tFL has been tested in very small series of patients, in which there has often been high transplant-related mortality (Table 2) (84,85,95,96,98-102). Whether a better outcome may be achieved by using reduced-intensity conditioning in contrast to the predominantly used myeloablative conditioning is unknown (96).

Patients with transformed lymphoma are often excluded from prospective clinical trials, so information about the efficacy of new drugs for treating tFL is scarce. Czuczman et al. reported a phase 2 study using the immunomodulatory agent lenalidomide that produced an overall response rate of 57%, and a median response duration of over 1 year in patients with tFL (106). Likewise, promising results were obtained from patients with histological transformation treated with radio-immunotherapy (82). More recently, a plethora of new drugs targeting crucial pathways in B-cell lymphomas has been developed, including PI3K pathway inhibitors (idelalisib), B-cell receptor inhibitors (ibrutinib), new anti-CD20 monoclonal antibodies (ofatumumab, obinutuzumab), BCL-2 inhibitors (venetoclax), immune-checkpoint inhibitors (e.g., PD1-PDL1/PDL2 pathway inhibitors nivolumab, pidilizumab, or pembrolizumab), and EZH2 inhibitors, among others (107-116). Some of these new molecules (e.g., ibrutinib, lenalidomide) have been shown to be more effective in a specific DLBCL subtype (113,117). Clinical trials of new drugs in tFL should include an analysis of the COO. Unfortunately, patients with transformed lymphoma have been excluded from most of these clinical trials. Some of the drugs (e.g., ibrutinib, idelalisib, venetoclax) revert the bad prognosis of TP53 alterations in different B-LPDs, providing a therapeutic option for these patients and, thereby, the possibility of reducing the risk of transformation (118).

In summary, increasing knowledge of the biology and treatment of FL and DLBCL patients, which translates into new therapeutic strategies, contrasts with the limited advances in or understanding of tFL. It is expected that a better knowledge of the biology of tFL will lead to well-designed clinical trials including or focusing on tFL.


Conclusions and remarks

The implementation of new tools has broadly improved our knowledge of the biology of NHL in general, and of tFL in particular. However, genetic studies on tFL are generally targeted to certain genes or regions. There is a lack of studies that evaluate the predictive role of multiple genes such as the m7-FLIPI for development of transformation. Moreover, most studies do not consider the variety of genetic alterations (CNAs, translocations, mutations) in the same cohort, or consider these along with the composition of the microenvironment, or other potential biological markers (SNPs, HLAs). Furthermore, the studied tFL cohorts have not distinguished whether transformation has presented as a first or subsequent event, which could misinterpret the role of certain gene alterations.

A comprehensive study integrating the results of the previous studies in large-scale series of clonally related FL-tFL sample pairs would help improve our knowledge of the developmental pathways leading to transformation. Consequently, it would be feasible to develop an index based on the most important biological and clinical factors that accurately stratify patients who will transform, distinguishing them from those who will relapse, since they require different therapeutic strategies. Furthermore, identifying patients who will never transform or progress will help individualize treatments in order to reduce toxicity and improve their quality-of-life.

Due to the limited availability of biopsy material for determining histological criteria, there is an unmet need to establish alternative but precise criteria to clearly discriminate transformation from FL progression. Effort needs to be directed towards this objective: a prospective study including clinical, biological, and imaging criteria would enable a specific and sensitive index to be developed. It would also allow the more accurate estimation of the real incidence of FL transformation, and provide an optimized definition of tFL.

The role of treatment strategies for tFL, including new agents, auto-SCT, and allo-SCT, must be determined from large-scale studies. To this end, common efforts of cooperative international groups (e.g., the ARISTOTLE project) could clearly help fulfill these unmet needs.

The hope is that, in the future, biological knowledge will enable personalized approaches to be developed to treat tFL, and/or patients with FL, in such a way as to avoid transformation or refractoriness.


Acknowledgments

Funding: This work was supported in part by grants from the Health Research Program of the Institute of Health Carlos III (ISCIII), the Spanish Ministry of Economy and Competitiveness, FIS-PI15/01393, CIBERONC (CB16/12/00233), the Education Counseling of Castilla y León (CAS102P17), and the European Regional Development Fund (ERDF) ‘Una manera de hacer Europa’ (Innocampus; CEI-2010-1-0010). All Spanish funding was co-sponsored by the European Union FEDER program.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Bruce D. Cheson) for the series “Inaugural Issue” published in Annals of Lymphoma. The article has undergone external peer review.

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/aol.2017.11.03). The series “Inaugural Issue” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Swerdlow SH, Campo E, Harris NL, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. International Agency for Research on Cancer. Lyon: IARC Press, 2008.
  2. Kridel R, Sehn LH, Gascoyne RD. Pathogenesis of follicular lymphoma. J Clin Invest 2012;122:3424-31. [Crossref] [PubMed]
  3. Hiddemann W, Kneba M, Dreyling M, et al. Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: results of a prospective randomized study of the German Low-Grade Lymphoma Study Group. Blood 2005;106:3725-32. [Crossref] [PubMed]
  4. Marcus R, Imrie K, Solal-Celigny P, et al. Phase III study of R-CVP compared with cyclophosphamide, vincristine, and prednisone alone in patients with previously untreated advanced follicular lymphoma. J Clin Oncol 2008;26:4579-86. [Crossref] [PubMed]
  5. Salles G, Seymour JF, Offner F, et al. Rituximab maintenance for 2 years in patients with high tumour burden follicular lymphoma responding to rituximab plus chemotherapy (PRIMA): a phase 3, randomised controlled trial. Lancet 2011;377:42-51. [Crossref] [PubMed]
  6. 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;33:2516-22. [Crossref] [PubMed]
  7. Sorigue M, Mercadal S, Alonso S, et al. Refractoriness to immunochemotherapy in follicular lymphoma: Predictive factors and outcome. Hematol Oncol 2017; [Epub ahead of print]. [Crossref] [PubMed]
  8. Magnano L, Alonso-Álvarez S, Alcoceba M, et al. Patients with follicular lymphoma (FL) in maintained complete response (CR) at 30 months show a survival similar to a sex- and age-matched Spanish general population. Hematol Oncol 2017;35:226-7. [Crossref]
  9. 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;25:2426-33. [Crossref] [PubMed]
  10. Casulo C, Burack WR, Friedberg JW. Transformed follicular non-Hodgkin lymphoma. Blood 2015;125:40-7. [Crossref] [PubMed]
  11. Alonso-Álvarez S, Magnano L, Alcoceba M, et al. Risk of and survival following histological transformation in follicular lymphoma in the rituximab era. A retrospective multicenter study by the Spanish GELTAMO group. Br J Haematol 2017;178:699-708. [Crossref] [PubMed]
  12. Kridel R, Sehn LH, Gascoyne RD. Can histologic transformation of follicular lymphoma be predicted and prevented? Blood 2017;130:258-66. [Crossref] [PubMed]
  13. Salles G, Coiffier B. Histologic transformation in follicular lymphoma. Ann Oncol 1998;9:803-5. [Crossref] [PubMed]
  14. Giné E, Montoto S, Bosch F, 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 2006;17:1539-45. [Crossref] [PubMed]
  15. Bastion Y, Sebban C, Berger F, et al. Incidence, predictive factors, and outcome of lymphoma transformation in follicular lymphoma patients. J Clin Oncol 1997;15:1587-94. [Crossref] [PubMed]
  16. 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;26:5165-9. [Crossref] [PubMed]
  17. 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/MayoClinic Specialized Program of Research Excellence Molecular Epidemiology Resource. J Clin Oncol 2013;31:3272-8. [Crossref] [PubMed]
  18. Lossos IS, Gascoyne RD. Transformation of follicular lymphoma. Best Pract Res Clin Haematol 2011;24:147-63. [Crossref] [PubMed]
  19. Schöder H, Noy A, Gonen M, et al. Intensity of 18fluorodeoxyglucose uptake in positron emission tomography distinguishes between indolent and aggressive non-Hodgkin's lymphoma. J Clin Oncol 2005;23:4643-51. [Crossref] [PubMed]
  20. Karam M, Novak L, Cyriac J, et al. Role of fluorine-18 fluoro-deoxyglucose positron emission tomography scan in the evaluation and follow-up of patients with low-grade lymphomas. Cancer 2006;107:175-83. [Crossref] [PubMed]
  21. Bodet-Milin C, Kraeber-Bodere F, Moreau P, et al. Investigation of FDG-PET/CT imaging to guide biopsies in the detection of histological transformation of indolent lymphoma. Haematologica 2008;93:471-2. [Crossref] [PubMed]
  22. Noy A, Schoder H, Gonen M, 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 2009;20:508-12. [Crossref] [PubMed]
  23. Ngeow JY, Quek RH, Ng DC, et al. High SUV uptake on FDG-PET/CT predicts for an aggressive B-cell lymphoma in a prospective study of primary FDG-PET/CT staging in lymphoma. Ann Oncol 2009;20:1543-7. [Crossref] [PubMed]
  24. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016;127:2375-90. [Crossref] [PubMed]
  25. Davies AJ, Rosenwald A, Wright G, et al. Transformation of follicular lymphoma to diffuse large B-cell lymphoma proceeds by distinct oncogenic mechanisms. Br J Haematol 2007;136:286-93. [Crossref] [PubMed]
  26. Kridel R, Mottok A, Farinha P, et al. Cell of origin of transformed follicular lymphoma. Blood 2015;126:2118-27. [Crossref] [PubMed]
  27. Al-Mansour M, Connors JM, Gascoyne RD, et al. Transformation to aggressive lymphoma in nodular lymphocyte-predominant Hodgkin's lymphoma. J Clin Oncol 2010;28:793-9. [Crossref] [PubMed]
  28. Rossi D, Gaidano G. Richter syndrome. Adv Exp Med Biol 2013;792:173-91. [Crossref] [PubMed]
  29. Conconi A, Franceschetti S, Aprile von Hohenstaufen K, et al. Histologic transformation in marginal zone lymphomas. Ann Oncol 2015;26:2329-35. [Crossref] [PubMed]
  30. Casulo C, Friedberg J. Transformation of marginal zone lymphoma (and association with other lymphomas). Best Pract Res Clin Haematol 2017;30:131-8. [Crossref] [PubMed]
  31. Jiménez C, Alonso-Álvarez S, Alcoceba M, et al. From Waldenstrom's macroglobulinemia to aggressive diffuse large B-cell lymphoma: a whole-exome analysis of abnormalities leading to transformation. Blood Cancer J 2017;7:e591 [Crossref] [PubMed]
  32. Conconi A, Ponzio C, Lobetti-Bodoni C, et al. Incidence, risk factors and outcome of histological transformation in follicular lymphoma. Br J Haematol 2012;157:188-96. [Crossref] [PubMed]
  33. Bains P, Al Tourah A, Campbell BA, et al. Incidence of transformation to aggressive lymphoma in limited-stage follicular lymphoma treated with radiotherapy. Ann Oncol 2013;24:428-32. [Crossref] [PubMed]
  34. 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;126:851-7. [Crossref] [PubMed]
  35. 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;34:2575-82. [Crossref] [PubMed]
  36. Federico M, Caballero D, Marcheselli L, et al. The risk of transformation of follicular lymphoma "transformed" by rituximab: the ARISTOTLE study promoted by the European lymphoma institute. Hematol Oncol 2017;35:115-6. [Crossref]
  37. Eide MB, Liestol K, Lingjaerde OC, et al. Genomic alterations reveal potential for higher grade transformation in follicular lymphoma and confirm parallel evolution of tumor cell clones. Blood 2010;116:1489-97. [Crossref] [PubMed]
  38. García-Álvarez M, Alonso-Álvarez S, Alcoceba M, et al. VDJH usage in transformed follicular lymphoma. Hematol Oncol 2015;33:244-321.
  39. Montoto S, Fitzgibbon J. Transformation of indolent B-cell lymphomas. J Clin Oncol 2011;29:1827-34. [Crossref] [PubMed]
  40. Garvin AJ, Simon RM, Osborne CK, et al. An autopsy study of histologic progression in non-Hodgkin's lymphomas. 192 cases from the National Cancer Institute. Cancer 1983;52:393-8. [Crossref] [PubMed]
  41. Delgado J, Pereira A, Villamor N, et al. Survival analysis in hematologic malignancies: recommendations for clinicians. Haematologica 2014;99:1410-20. [Crossref] [PubMed]
  42. Kridel R, Chan FC, Mottok A, et al. Histological Transformation and Progression in Follicular Lymphoma: A Clonal Evolution Study. PLoS Med 2016;13:e1002197 [Crossref] [PubMed]
  43. Mozessohn L, Cheung MC, Crump M, et al. Chemoimmunotherapy resistant follicular lymphoma: predictors of resistance, association with transformation and prognosis. Leuk Lymphoma 2014;55:2502-7. [Crossref] [PubMed]
  44. Rossi D, Ciardullo C, Spina V, et al. Molecular bases of chronic lymphocytic leukemia in light of new treatments. Immunol Lett 2013;155:51-5. [Crossref] [PubMed]
  45. 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;15:424-35. [Crossref] [PubMed]
  46. Morin RD, Mendez-Lago M, Mungall AJ, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 2011;476:298-303. [Crossref] [PubMed]
  47. Lohr JG, Stojanov P, Lawrence MS, et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc Natl Acad Sci U S A 2012;109:3879-84. [Crossref] [PubMed]
  48. Morin RD, Mungall K, Pleasance E, et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 2013;122:1256-65. [Crossref] [PubMed]
  49. Okosun J, Bodor C, Wang J, et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet 2014;46:176-81. [Crossref] [PubMed]
  50. Pasqualucci L, Khiabanian H, Fangazio M, et al. Genetics of follicular lymphoma transformation. Cell Rep 2014;6:130-40. [Crossref] [PubMed]
  51. O'Shea D, O’Riain C, Taylor C, et al. The presence of TP53 mutation at diagnosis of follicular lymphoma identifies a high-risk group of patients with shortened time to disease progression and poorer overall survival. Blood 2008;112:3126-9. [Crossref] [PubMed]
  52. Cheung KJ, Johnson NA, Affleck JG, et al. Acquired TNFRSF14 mutations in follicular lymphoma are associated with worse prognosis. Cancer Res 2010;70:9166-74. [Crossref] [PubMed]
  53. Trinh DL, Scott DW, Morin RD, et al. Analysis of FOXO1 mutations in diffuse large B-cell lymphoma. Blood 2013;121:3666-74. [Crossref] [PubMed]
  54. Fernández-Rodríguez C, Bellosillo B, Garcia-Garcia M, et al. MYD88 (L265P) mutation is an independent prognostic factor for outcome in patients with diffuse large B-cell lymphoma. Leukemia 2014;28:2104-6. [Crossref] [PubMed]
  55. Pastore A, Jurinovic V, Kridel R, et al. Integration of gene mutations in risk prognostication for patients receiving first-line immunochemotherapy for follicular lymphoma: a retrospective analysis of a prospective clinical trial and validation in a population-based registry. Lancet Oncol 2015;16:1111-22. [Crossref] [PubMed]
  56. Jurinovic V, Kridel R, Staiger AM, et al. Clinicogenetic risk models predict early progression of follicular lymphoma after first-line immunochemotherapy. Blood 2016;128:1112-20. [Crossref] [PubMed]
  57. Bouska A, Zhang W, Gong Q, et al. Combined copy number and mutation analysis identifies oncogenic pathways associated with transformation of follicular lymphoma. Leukemia 2017;31:83-91. [Crossref] [PubMed]
  58. Skibola CF, Bracci PM, Halperin E, et al. Genetic variants at 6p21.33 are associated with susceptibility to follicular lymphoma. Nat Genet 2009;41:873-5. [Crossref] [PubMed]
  59. Conde L, Halperin E, Akers NK, et al. Genome-wide association study of follicular lymphoma identifies a risk locus at 6p21.32. Nat Genet 2010;42:661-4. [Crossref] [PubMed]
  60. Smedby KE, Foo JN, Skibola CF, et al. GWAS of follicular lymphoma reveals allelic heterogeneity at 6p21.32 and suggests shared genetic susceptibility with diffuse large B-cell lymphoma. PLoS Genet 2011;7:e1001378 [Crossref] [PubMed]
  61. Chaperot L, Delfau-Larue MH, Jacob MC, et al. Differentiation of antitumor-specific cytotoxic T lymphocytes from autologous tumor infiltrating lymphocytes in non-Hodgkin's lymphomas. Exp Hematol 1999;27:1185-93. [Crossref] [PubMed]
  62. Wang SS, Abdou AM, Morton LM, et al. Human leukocyte antigen class I and II alleles in non-Hodgkin lymphoma etiology. Blood 2010;115:4820-3. [Crossref] [PubMed]
  63. Akers NK, Curry JD, Conde L, et al. Association of HLA-DQB1 alleles with risk of follicular lymphoma. Leuk Lymphoma 2011;52:53-8. [Crossref] [PubMed]
  64. Lu Y, Abdou AM, Cerhan JR, et al. Human leukocyte antigen class I and II alleles and overall survival in diffuse large B-cell lymphoma and follicular lymphoma. ScientificWorldJournal 2011;11:2062-70. [Crossref] [PubMed]
  65. Alcoceba M, Sebastian E, Marin L, et al. HLA specificities are related to development and prognosis of diffuse large B-cell lymphoma. Blood 2013;122:1448-54. [Crossref] [PubMed]
  66. Wrench D, Leighton P, Skibola CF, et al. SNP rs6457327 in the HLA region on chromosome 6p is predictive of the transformation of follicular lymphoma. Blood 2011;117:3147-50. [Crossref] [PubMed]
  67. Berglund M, Enblad G, Thunberg U. SNP rs6457327 is a predictor for overall survival in follicular lymphoma as well as survival after transformation. Blood 2011;118:4489. [Crossref] [PubMed]
  68. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med 2015;372:311-9. [Crossref] [PubMed]
  69. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189-99. [Crossref] [PubMed]
  70. Farinha P, Al-Tourah A, Gill K, et al. The architectural pattern of FOXP3-positive T cells in follicular lymphoma is an independent predictor of survival and histologic transformation. Blood 2010;115:289-95. [Crossref] [PubMed]
  71. Glas AM, Knoops L, Delahaye L, 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 2007;25:390-8. [Crossref] [PubMed]
  72. Farinha P, Masoudi H, Skinnider BF, et al. Analysis of multiple biomarkers shows that lymphoma-associated macrophage (LAM) content is an independent predictor of survival in follicular lymphoma (FL). Blood 2005;106:2169-74. [Crossref] [PubMed]
  73. Alvaro T, Lejeune M, Camacho FI, et al. The presence of STAT1-positive tumor-associated macrophages and their relation to outcome in patients with follicular lymphoma. Haematologica 2006;91:1605-12. [PubMed]
  74. de Jong D, Koster A, Hagenbeek A, et al. Impact of the tumor microenvironment on prognosis in follicular lymphoma is dependent on specific treatment protocols. Haematologica 2009;94:70-7. [Crossref] [PubMed]
  75. Taskinen M, Karjalainen-Lindsberg ML, Nyman H, et al. A high tumor-associated macrophage content predicts favorable outcome in follicular lymphoma patients treated with rituximab and cyclophosphamide-doxorubicin-vincristine-prednisone. Clin Cancer Res 2007;13:5784-9. [Crossref] [PubMed]
  76. Canioni D, Salles G, Mounier N, et al. High numbers of tumor-associated macrophages have an adverse prognostic value that can be circumvented by rituximab in patients with follicular lymphoma enrolled onto the GELA-GOELAMS FL-2000 trial. J Clin Oncol 2008;26:440-6. [Crossref] [PubMed]
  77. Taskinen M, Karjalainen-Lindsberg ML, Leppa S. Prognostic influence of tumor-infiltrating mast cells in patients with follicular lymphoma treated with rituximab and CHOP. Blood 2008;111:4664-7. [Crossref] [PubMed]
  78. Farinha P, Kyle AH, Minchinton AI, et al. Vascularization predicts overall survival and risk of transformation in follicular lymphoma. Haematologica 2010;95:2157-60. [Crossref] [PubMed]
  79. Fu K, Weisenburger DD, Choi WW, et al. Addition of rituximab to standard chemotherapy improves the survival of both the germinal center B-cell-like and non-germinal center B-cell-like subtypes of diffuse large B-cell lymphoma. J Clin Oncol 2008;26:4587-94. [Crossref] [PubMed]
  80. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 2002;346:235-42. [Crossref] [PubMed]
  81. Pfreundschuh M, Kuhnt E, Trumper L, et al. CHOP-like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomised study of the MabThera International Trial (MInT) Group. Lancet Oncol 2011;12:1013-22. [Crossref] [PubMed]
  82. Kaminski MS, Estes J, Zasadny KR, et al. Radioimmunotherapy with iodine (131)I tositumomab for relapsed or refractory B-cell non-Hodgkin lymphoma: updated results and long-term follow-up of the University of Michigan experience. Blood 2000;96:1259-66. [PubMed]
  83. 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;58:1805-13. [Crossref] [PubMed]
  84. 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;163:487-95. [Crossref] [PubMed]
  85. 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;31:1164-71. [Crossref] [PubMed]
  86. 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;26:393-9. [Crossref] [PubMed]
  87. Foran JM, Apostolidis J, Papamichael D, et al. High-dose therapy with autologous haematopoietic support in patients with transformed follicular lymphoma: a study of 27 patients from a single centre. Ann Oncol 1998;9:865-9. [Crossref] [PubMed]
  88. Williams CD, Harrison CN, Lister TA, et al. High-dose therapy and autologous stem-cell support for chemosensitive transformed low-grade follicular non-Hodgkin's lymphoma: a case-matched study from the European Bone Marrow Transplant Registry. J Clin Oncol 2001;19:727-35. [Crossref] [PubMed]
  89. Andreadis C, Schuster SJ, Chong EA, et al. Long-term event-free survivors after high-dose therapy and autologous stem-cell transplantation for low-grade follicular lymphoma. Bone Marrow Transplant 2005;36:955-61. [Crossref] [PubMed]
  90. Sabloff M, Atkins HL, Bence-Bruckler I, et al. A 15-year analysis of early and late autologous hematopoietic stem cell transplant in relapsed, aggressive, transformed, and nontransformed follicular lymphoma. Biol Blood Marrow Transplant 2007;13:956-64. [Crossref] [PubMed]
  91. Hamadani M, Benson DM Jr, Lin TS, et al. High-dose therapy and autologous stem cell transplantation for follicular lymphoma undergoing transformation to diffuse large B-cell lymphoma. Eur J Haematol 2008;81:425-31. [Crossref] [PubMed]
  92. 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;50:1625-31. [Crossref] [PubMed]
  93. Eide MB, Lauritzsen GF, Kvalheim G, et al. High dose chemotherapy with autologous stem cell support for patients with histologically transformed B-cell non-Hodgkin lymphomas. A Norwegian multi centre phase II study. Br J Haematol 2011;152:600-10. [Crossref] [PubMed]
  94. 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;53:830-5. [Crossref] [PubMed]
  95. 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;12:406-11. [Crossref] [PubMed]
  96. 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;20:951-9. [Crossref] [PubMed]
  97. Kuruvilla J, MacDonald DA, Kouroukis CT, et al. Salvage chemotherapy and autologous stem cell transplantation for transformed indolent lymphoma: a subset analysis of NCIC CTG LY12. Blood 2015;126:733-8. [Crossref] [PubMed]
  98. Rezvani AR, Storer B, Maris M, et al. Nonmyeloablative allogeneic hematopoietic cell transplantation in relapsed, refractory, and transformed indolent non-Hodgkin's lymphoma. J Clin Oncol 2008;26:211-7. [Crossref] [PubMed]
  99. Hamadani M, Awan FT, Elder P, et al. Feasibility of allogeneic hematopoietic stem cell transplantation for follicular lymphoma undergoing transformation to diffuse large B-cell lymphoma. Leuk Lymphoma 2008;49:1893-8. [Crossref] [PubMed]
  100. Ramadan KM, Connors JM, Al-Tourah AJ, et al. Allogeneic SCT for relapsed composite and transformed lymphoma using related and unrelated donors: long-term results. Bone Marrow Transplant 2008;42:601-8. [Crossref] [PubMed]
  101. Thomson KJ, Morris EC, Bloor A, et al. Favorable long-term survival after reduced-intensity allogeneic transplantation for multiple-relapse aggressive non-Hodgkin's lymphoma. J Clin Oncol 2009;27:426-32. [Crossref] [PubMed]
  102. Clavert A, Le GS, Brissot E, et al. Reduced-intensity conditioning allogeneic stem cell transplant for relapsed or transformed aggressive B-cell non-Hodgkin lymphoma. Leuk Lymphoma 2010;51:1502-8. [Crossref] [PubMed]
  103. Sorigue M, Garcia O, Baptista MJ, et al. Similar prognosis of transformed and de novo diffuse large B-cell lymphomas in patients treated with immunochemotherapy. Med Clin (Barc) 2017;148:243-9. [Crossref] [PubMed]
  104. Montoto S. Treatment of patients with transformed lymphoma. Hematology Am Soc Hematol Educ Program 2015;2015:625-30.
  105. 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;333:1540-5. [Crossref] [PubMed]
  106. Czuczman MS, Vose JM, Witzig TE, et al. The differential effect of lenalidomide monotherapy in patients with relapsed or refractory transformed non-Hodgkin lymphoma of distinct histological origin. Br J Haematol 2011;154:477-81. [Crossref] [PubMed]
  107. Gopal AK, Kahl BS, de Vos S, et al. PI3Kdelta inhibition by idelalisib in patients with relapsed indolent lymphoma. N Engl J Med 2014;370:1008-18. [Crossref] [PubMed]
  108. Younes A, Thieblemont C, Morschhauser F, et al. Combination of ibrutinib with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) for treatment-naive patients with CD20-positive B-cell non-Hodgkin lymphoma: a non-randomised, phase 1b study. Lancet Oncol 2014;15:1019-26. [Crossref] [PubMed]
  109. Maddocks K, Christian B, Jaglowski S, et al. A phase 1/1b study of rituximab, bendamustine, and ibrutinib in patients with untreated and relapsed/refractory non-Hodgkin lymphoma. Blood 2015;125:242-8. [Crossref] [PubMed]
  110. Davids MS, Roberts AW, Seymour JF, et al. Phase I First-in-Human Study of Venetoclax in Patients With Relapsed or Refractory Non-Hodgkin Lymphoma. J Clin Oncol 2017;35:826-33. [Crossref] [PubMed]
  111. Hiddemann W, Barbui AM, Canales Albendea MA, et al. Immunochemotherapy with obinutuzumab or rituximab in previously untreated follicular lymphoma in the randomised phase III GALLIUM study: analysis by chemotherapy regimen. Hematol Oncol 2017;35:117-9. [Crossref]
  112. Westin JR, Chu F, Zhang M, et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol 2014;15:69-77. [Crossref] [PubMed]
  113. 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;33:251-7. [Crossref] [PubMed]
  114. Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J Clin Oncol 2016;34:2698-704. [Crossref] [PubMed]
  115. Nastoupil LJ, Westin J, Fowler N, et al. High response rates with pembrolizumab in combination with rituximab in patients with relapsed follicular lymphoma: interim results of an on open-label, phase II study. Hematol Oncol 2017;35:121-2. [Crossref]
  116. McCabe MT, Ott HM, Ganji G, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 2012;492:108-12. [Crossref] [PubMed]
  117. Wilson WH, Young RM, Schmitz R, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med 2015;21:922-6. [Crossref] [PubMed]
  118. Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med 2013;369:32-42. [Crossref] [PubMed]
doi: 10.21037/aol.2017.11.03
Cite this article as: Alcoceba M, Alonso-Álvarez S, García-Álvarez M, Martín A, Caballero MD. Unmet needs in histological transformation of follicular lymphoma: a clinical and biological review. Ann Lymphoma 2017;1:11.

Download Citation