Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Apr 24, 2023; 14(4): 160-170
Published online Apr 24, 2023. doi: 10.5306/wjco.v14.i4.160
Integration of molecular testing for the personalized management of patients with diffuse large B-cell lymphoma and follicular lymphoma
Ruth Stuckey, Hugo Luzardo Henríquez, Haridian de la Nuez Melian, Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas 35019, Spain
José Carlos Rivero Vera, Department of Anatomical Pathology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas 35019, Spain
Cristina Bilbao-Sieyro, María Teresa Gómez-Casares, Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrin, Las Palmas de Gran Canaria 35019, Las Palmas de Gran Canaria, Spain
Cristina Bilbao-Sieyro, Department of Morphology, Universitario de Las Palmas de Gran Canaria, Las Palmas 35001, Spain
María Teresa Gómez-Casares, Medical Science, Universitario de Las Palmas de Gran Canaria, Las Palmas 35001, Spain
ORCID number: Ruth Stuckey (0000-0001-6955-2290); Cristina Bilbao-Sieyro (0000-0002-4796-1445); María Teresa Gómez-Casares (0000-0003-0505-5126).
Author contributions: Stuckey R performed the majority of the writing, and prepared the tables; Luzardo Henríquez H, de la Nuez Melian H and Rivero Vera JC provided input in writing the paper; Bilbao-Sieyro C and Gómez-Casares MT coordinated the writing of the paper; all authors approved the final version of the manuscript.
Conflict-of-interest statement: All the authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Cristina Bilbao-Sieyro, PhD, Lecturer, Technician, Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrin, Barranco de la Ballena, Las Palmas de Gran Canaria 35019, Las Palmas de Gran Canaria, Spain. bilbaocristina@gmail.com
Received: December 21, 2022
Peer-review started: December 21, 2022
First decision: March 24, 2023
Revised: March 24, 2023
Accepted: April 4, 2023
Article in press: April 4, 2023
Published online: April 24, 2023

Abstract

Diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) are the most common forms of aggressive and indolent lymphoma, respectively. The majority of patients are cured by standard R-CHOP immunochemotherapy, but 30%–40% of DLBCL and 20% of FL patients relapse or are refractory (R/R). DLBCL and FL are phenotypically and genetically hereterogenous B-cell neoplasms. To date, the diagnosis of DLBCL and FL has been based on morphology, immunophenotyping and cytogenetics. However, next-generation sequencing (NGS) is widening our understanding of the genetic basis of the B-cell lymphomas. In this review we will discuss how integrating the NGS-based characterization of somatic gene mutations with diagnostic or prognostic value in DLBCL and FL could help refine B-cell lymphoma classification as part of a multidisciplinary pathology work-up. We will also discuss how molecular testing can identify candidates for clinical trials with targeted therapies and help predict therapeutic outcome to currently available treatments, including chimeric antigen receptor T-cell, as well as explore the application of circulating cell-free DNA, a non-invasive method for patient monitoring. We conclude that molecular analyses can drive improvements in patient outcomes due to an increased understanding of the different pathogenic pathways affected by each DLBCL subtype and indolent FL vs R/R FL.

Key Words: Next-generation sequencing, Prognosis, Molecular analysis, Targeted therapy, Chimeric antigen receptor T-cell therapy, Personalized medicine

Core Tip: Molecular studies in the past decade have improved our understanding of the biological heterogeneity of B-cell lymphomas such as diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. Next-generation sequencing studies are helping to reveal the different pathogenic pathways affected by each DLBCL subtype and identify new targets for directed therapy. Molecular analysis can also help predict therapeutic outcome to currently available treatments, including chimeric antigen receptor T-cell therapy, and identify candidates for clinical trials with targeted therapies, ultimately leading to improvements in patient outcomes. As such, the incorporation of precision medicine via the integration of molecular analyses in clinical practice can improve clinical outcomes in patients and thus contribute to a new standard of care for patients with B-cell lymphomas.



INTRODUCTION

B-cell lymphomas are classified into over 19 distinct entities, as defined by the 2022 World Health Organization (WHO) classification[1]. Diffuse large B-cell lymphoma (DLBCL) is the most common form of non-Hodgkin lymphoma (NHL), representing approximately 30% of lymphomas of mature B-cells[2], while follicular lymphoma (FL) is the second most common NHL. However, both DLBCL and FL are phenotypically and genetically hereterogenous B-cell neoplasms. For example, the majority of DLBCL patients are cured by standard rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone (R-CHOP) immunochemotherapy, but 30%–40% relapse or are refractory (R/R), while for FL, approximately 20% of patients treated with chemoimmunotherapy will progress within the first two years of diagnosis (POD24)[3]. Thus, improvements in patient outcomes will rely on an increased understanding of the different pathogenic DLBCL and FL pathways that lead to treatment failure and/or progression.

Next-generation sequencing (NGS) studies together with copy-number analysis are determining genes with recurrent alterations in DLBCL and FL, some of which can refine diagnosis and prognostic stratification. In this minireview, we will describe how molecular analyses are revealing differences in somatic mutations according to disease subtypes, helping with differential diagnosis, as well as determining new targets for the development of directed therapies. We will also explore the application of circulating cell-free DNA, a non-invasive method for patient monitoring. Finally, we will discuss how the incorporation of precision medicine can identify candidates for clinical trials with targeted therapies and help predict therapeutic outcome to currently available treatments in a drive towards a more personalized treatment approach.

We aim to convince the reader that the incorporation of molecular testing for somatic gene mutations can improve the diagnosis and prognosis of patients with DLBCL and FL as part of a multidisciplinary pathology work-up.

CONVENTIONAL CLASSIFICATION OF DLBCL

Currently, using immunohistochemistry (IHC), fluorescence in situ hybridization (FISH) and flow cytometry techniques, patients with DLBCL are divided into three subtypes depending on the stage of differentiation and maturation of the B cells of origin: germinal center B-cell like (GCB) or activated B-cell like (ABC), with the remaining 10% to 20% “unclassified” or not otherwise specified (NOS). For example, IHC analysis of CD10, BCL6 and MUM1 markers helps determine the GCB and ABC subtypes according to the Hans algorithm[4]. Nevertheless, the Hans algorithm doesn’t distinguish the NOS DLBCL subtype and gives an incorrect classification in approximately 20% of cases[4].

DLBCL patients show distinct clinical outcomes according to the subtype: patients with the GCB phenotype have a more favorable outcome than ABC in terms of survival when treated with standard chemotherapy, 60% at 5 years vs 35% in ABC[5]. In addition, IHC of MYC and BCL2/BCL6 can identify tumors as double- or triple-expressor, associated with worse prognosis[6]. Studies involving FISH analysis of MYC rearrangements have shown that MYC rearranged with an immunoglobulin (IG) gene has worse prognosis compared to MYC with a non-IG partner, with MYC/IG double hits associated with an even poorer prognosis[7]. Thus, genomic tests used in routine clinical practice are already adding prognostic value. Even so, the diagnostic work-up and treatment are practically identical for all DLBCL patients despite the high genetic heterogeneity.

In terms of treatment, R-CHOP has been the standard of care for over two decades, and is still administered to the majority of DLBCL patients[8]. To improve treatment response, and predict which patients are likely to be R/R, elucidation of the molecular determinants related to treatment response will be fundamental. One advancement in this area is the observation that high EZH2 expression (> 70%, detected by IHC) is associated with superior survival of DLBCL patients following R-CHOP[9].

CONVENTIONAL CLASSIFICATION OF FL

FL is characterized by the t(14;18)(q32;q21) translocation, present in 90% of FL patients, resulting in overexpression of BCL2 under the IGH promoter. In cases lacking t(14;18), BCL6 and CD10 expression patterns confirm FL diagnosis[10]. Rearrangement of BCL6 (3q27) may also be found in grade 3 FL, with or without t(14;18)[11].

Prognostic biological factors include age > 60 years and hemoglobin < 12 g/dL, as well as other biomarkers, such as LDH or β2-microglobin above normal, according to the FLIPI and FLIPI-2 scores, respectively[12,13].

Several first-lines of immunochemotherapy exist, including bendamustine + rituximab, rituximab alone, or R-CHOP, with choice largely down to the clinician’s preference. Treatment improvements are a necessity, given that POD24 is a predictor of overall survival (OS), with rates of just 50% for patients with POD24 vs 90% in those with no POD24 following R-CHOP treatment[3]. As highlighted in the recent editorial by Leonard[14], there is currently “no reliable way” to determine at diagnosis whether a patient with FL is likely to respond optimally to immunochemotherapy. The hope is that molecular analyses could help identify a subgroup of at-risk patients who would benefit from upfront treatment with a specific targeted therapy.

NGS APPLICATION IN LYMPHOMAS

According to the 2022 WHO and ICC classifications and the European Society for Medical Oncology’s 2021 clinical guidelines, no molecular analyses are currently recommended at diagnosis for DLBCL or FL[1,15,16]. To date, only a few entities of lymphoid neoplasms are defined by genomic criteria. This is in stark contrast to other hematological malignancies, in particular myeloid neoplasms, where the use of NGS is well-established in diagnosis and risk-stratification[1,16,17]. For example, for acute myeloid leukemia a complete genomic evaluation, including NGS panel, is obligatory at diagnosis to define disease subtypes and to direct therapies[1,16,17]. Nevertheless, both international consortiums acknowledge that molecular analyses in B-cell lymphomas have identified genomic alterations “with diagnostic, prognostic, and predictive impact in different entities”[18] and explicitly state that it is highly probable that more entities will be defined by genomic criteria in the near future[1,16,18].

MOLECULAR ANALYSES IN DLBCL

In recent years, advances in next-generation sequencing (NGS) techniques are redefining our understanding of the genetic basis of lymphomas. Molecular studies are revealing recurrent genetic events and thus are helping to identify the key pathways that are important in DLBCL pathogenicity and evolution, and may even have prognostic impact[19].

Mutations in the genes MYD88, CARD11, EZH2 and CD79A/CD79B have been identified in approximately 40% of DLBCL and are considered drivers of lymphomagenesis[20]. Moreover, NGS studies have revealed that GCB and ABC have a distinct profile of somatic mutations. For instance, mutations in GNA13 are found in GCB but are rare in other B-cell lymphoma subtypes[21], whereas the MYD88 L265P mutation is found in ABC but is rarely identified in GCB DLBCL[21]. Thus, mutational information can assist in providing an accurate diagnosis, for example for the differential diagnosis of DLBCL from primary mediastinal large B-cell lymphoma (PMBCL, a relatively rare NHL with large B-cell morphology)[22], mantle cell lymphoma, or grade 3 FL. In addition, relapse has been associated with mutations in certain genes, such as the B2M and CD58 immune surveillance genes[23].

Recent findings suggest that tumor genotype also influences treatment response. For example, whole genome sequencing analysis of 20 patients with high-risk GCB DLBCL revealed that those with cryptic rearrangements of MYC or BCL2 (not detectable by FISH) had worse outcomes to R-CHOP[24]. Moreover, ABC tumors that harboured both a mutation in CD79B and the MYD88 L265P mutation were more sensitive to the BTK inhibitor ibrutinib, whereas NOS subtype tumors with the MYD88 L265P mutation and CD79B wild-type showed a poor response to ibrutinib[25].

Although NGS of DLBCL is not currently recommended in routine clinical practice[1,16], huge efforts are underway to characterize the prognostic value and thus functional impact of driver mutations, for instance via the whole-exome sequencing of 1001 DLBCL samples[26].

Such large-scale studies using NGS techniques together with copy-number analysis, to identify genes with recurrent alterations with prognostic value, have led to the proposition of new DLBCL classifications. After studying 574 DLBCL biopsy samples Schmitz et al[27] proposed four genetic subtypes termed MCD (based on the co-occurrence of MYD88 and CD79B mutations), BN2 (based on BCL6 fusions and NOTCH2 mutations), N1 (based on NOTCH1 mutations), and EZB (based on EZH2 mutations and BCL2 translocations). These subtypes differed in their responses to immunochemotherapy, with favorable survival in the BN2 and EZB subtypes and inferior outcomes in the MCD and N1 subtypes[27], whereas MCD and N1 subtypes responded well to R-CHOP with ibrutinib[28]. Importantly, data on the BN2 subtype, with overlap with the NOS subgroup, revealed that patients are likely to be responsive to antagonists of B-cell receptor signaling such as the BTK inhibitors. Similarly, Chapuy et al[29] studied 304 DLBCL biopsies and identified six genetic subgroups. Of note, mutations in CD79B were associated with relapse independently of the subtype or International Prognostic Index (IPI) risk group[30].

Table 1 Frequent mutations detected in follicular lymphoma. Adapted from[32,37].
Gene
Frequency in FL, %
KMT2D82
CREBBP65-70
HIST1H1C and/or HIST1H1E28
EZH220-25
EP30014
STAT612
CARD1111
TNFAIP311
SOCS18
TP536
MOLECULAR ANALYSES IN FL

Mutations in genes encoding epigenetic modifiers (and the resultant pattern of aberrant DNA methylation) are a molecular hallmark of FL (Table 1)[31,32]. Moreover, such mutations are likely to be early driver events[33].

NGS studies have revealed that the acquisition of additional mutations contributes to disease progression and the risk of transformation of FL to DLBCL. For example, TP53 mutations have been associated with shorter progression-free survival and OS[34,35], while the gain of mutations in genes such as EBF1, MYD88 and TNFAIP3 are associated with progression to a more aggressive disease[32]. Additionally, expression studies have revealed chromosome regions, such as 1p36 and 6q21 deletion associated with transformation[36]. Thus, genetic analyses can improve the prognostication of patients with FL[37].

In the case of FL, the mutational status of seven genes (EZH2, ARID1A, MEF2B, EP300, FOXO1, CREBBP and CARD11) was added to the preexisting Eastern Cooperative Oncology Group (ECOG) performance status, FL International Prognostic Index (ECOG PS and FLIPI) risk stratification algorithms to develop the m7-FLIPI risk score[38]. Application of the m7-FLIPI risk score defined a high-risk group with a significantly shorter failure-free survival after receiving first-line R-CHOP.

Specifically, mutations in EZH2 were associated with the low-risk m7-FLIPI group and with higher OS[38]. As such, the presence or absence of the EZH2 Y646 point mutation can help decide the chemotherapy regime in a patient-specific manner, since patients with such a mutation were shown to respond well to R-CHOP[38] (higher OS and lower relapse rate) while patients without this mutation responded better to bendamustine[39].

Although the m7-FLIPI was not prognostic for FL patients who received rituximab, patients with EZH2 mutations had longer time to treatment failure while EP300 mutations were associated with shorter time to treatment failure[40]. Therefore, it remains to be determined if the m7-FLIPI risk score is prognostic for FL patients treated with other chemotherapy regimes other than R-CHOP. Furthermore, the use of such risk scores in the routine clinical practice is not common, partly due to lack of availability of mutational studies in some centers[41].

TARGETED THERAPIES IN DLBCL

It is clear that improvements in DLBCL outcomes will rely on an increased understanding of the different pathogenic pathways affected by each DLBCL subtype. Indeed, an in silico drug discovery analysis showed that 46% of cases harbored at least one genomic alteration considered to be a potential drug response target (according to early clinical trials or preclinical assays in DLBCL or other B-cell lymphomas)[30].

But, to date, only one targeted therapy against a molecular driver has been approved for DLBCL – selinexor – although many others are in development. Selinexor is a specific inhibitor of the XPO1 nuclear export transporter protein that was approved by the FDA in June 2020 for the treatment of adults with R/R DLBCL NOS, including DLBCL progressed from FL, after at least two previous lines of therapy[42].

Besides targeted agents, many immunotherapy strategies are in development, with the aim to promote the immune recognition and cytotoxic attack of T cells or macrophages. Some have achieved approval, such as rituximab, a monoclonal antibody against the common surface antigen CD20, and polatuzumab vedotin (Pola; Polivy™), an antibody-drug conjugate. Pola includes an anti-CD79B monoclonal antibody for cell targeting, which upon binding allows the antineoplastic agent monomethyl auristatin E to enter the cell and inhibit microtubule assembly, preventing cell mitosis and ultimately causing apoptosis[43]. Pola was approved by the FDA in June 2019 in combination with rituximab and bendamustine for the treatment of adults with R/R DLBCL after at least two previous lines of therapy. Such strategies have the advantage that the surface antigens they target are universally expressed on all DLBCL subtypes.

TARGETED THERAPIES IN FL

Genetic studies have identified epigenetic mechanisms in the pathogenesis of both DLBCL and FL, such as acetylation/deacetylation affected by CREBBP and EP300 mutations, or histone methylation changes affected by EZH2 mutations. Indeed, Tazemetostat (Tazverik™), an EZH2 inhibitor, was the first directed therapy to be approved by the FDA (in June 2020) for the treatment of R/R FL after two lines of previous therapy[44]. The EZH2 mutation is predictive of Tazemetostat response but, interestingly, this targeted agent was also shown to improve the outcome of patients without an EZH2 mutation[44].

Other FDA-approved agents for R/R FL include four PI3K signaling inhibitors: Idelalisib (July 2014), copanlisib (September 2017), duvelisib (September 2018), and umbralisib (February 2021)[45-48]. Further information on FL therapies in development can be found in this recent review[37].

CAR-T

Besides targeted therapies, an improved understanding of the genetic and immune biology of DLBCL and FL has led to the development of chimeric antigen receptor T-cell (CAR-T) therapies, considered a major scientific breakthrough and offering an alternative treatment option for patients with R/R B-cell lymphomas[49,50].

In 2020, our center obtained the license to provide the European Medicine Agency-approved anti-CD19 CAR-T axicabtagene ciloleucel (Yescarta™) and Tisagenlecleucel (Kymriah™) for the treatment of adult patients with R/R DLBCL or PMBCL after two or more previous lines of treatment. As of February 2021, the third CAR-T lisocabtagene maraleucel (Breyanzi™) also obtained FDA approval for the treatment of R/R DLBCL[51].

CAR-T is also an option for the treatment of adult R/R FL patients after two or more previous lines of treatment[52,53], following the FDA approval of axicabtagene ciloleucel in March 2021.

The proliferation and persistence of CAR-T cells in the body is an important factor influencing therapy durability, with the loss of a CAR-T signal associated with progression of the disease[54]. A quantitative TaqMan PCR (qPCR) assay can be used to monitor the number of CAR-T cells circulating in peripheral blood via detection of the quimeric CD19 recognition domain (FMC63)[55]. Flow cytometry is an alternative method for CAR-T cell monitoring, but has the disadvantage that it needs to be carried out on fresh samples and has lower sensitivity. Future studies are required to explore the correlation between the expansion/persistence of CAR-T cells and clinical outcomes including treatment efficacy and clinical symptoms.

CIRCULATING CELL-FREE TUMOR DNA

Surgical excision biopsies are the gold-standard technique used in the diagnosis and follow-up of patients with lymphomas, although core needle biopsies are a useful and viable alternative under certain conditions[56]. However, both surgical excision and core needle biopsies are resource intensive, can be painful, and impact negatively on patients, and surgical excision biopsies, in particular, have an associated risk of morbidity due to bleeding and infection. Additionally, some lymphomas may not be easily accessible which can limit the availability of tissue for genomic studies. Moreover, the extraction of genomic DNA from formalin-fixed, paraffin-embedded biopsies for downstream NGS applications is not ideal since chemicals used in the fixation can degrade nucleic acids, thus decreasing NGS sensitivity.

Liquid biopsy techniques are currently being explored as non-invasive methods for tumor diagnosis and disease monitoring[57]. Circulating cell-free DNA (ctDNA), consisting of highly fragmented DNA in plasma that is released by normal or tumor cells that undergo apoptosis or necrosis[58], may better reflect intratumoral heterogeneity than can be obtained from a single tissue biopsy. Indeed, in comparison with the sequencing of genomic DNA extracted from the diagnostic tissue biopsy, the sequencing of ctDNA can identify somatic mutations with a similar accuracy and identified additional clinically relevant mutations that were not detected in the diagnostic tissue biopsy[59]. Moreover, the analysis of ctDNA could overcome some other limitations of biopsies. For example, in the case of a biopsy at an extranodal site, it is not uncommon for the paraffin block to also contain other non-tumoral tissue.

Due to their easy accessibility through non-invasive procedures (such as a simple peripheral blood draw), ctDNA analyses can be repeated regularly to track lymphomas over time, such as to monitor treatment response. Indeed, studies have shown that changes in ctDNA quantification correlated with positive responses to chemotherapy and could even detect relapse, months earlier than conventional CT scan monitoring[60]. Thus they may also be useful as “surveillance” methods in patients who have completed treatment but may be at risk of relapse, e.g. those with mutations in CD79B or those with a high pretreatment ctDNA quantitative burden for early relapse detection[32,59].

Future studies are required to optimize the application of ctDNA analyses in the management of patients with B-cell lymphomas. Nevertheless, ctDNA is currently used in the clinic in some fields of oncology, such as in the molecular profiling of patients with non-squamous non-small cell lung cancer at diagnosis, as recommended by the National Comprehensive Cancer Network[61].

IMPLEMENTATION OF MOLECULAR TESTING IN CLINICAL PRACTICE

The application of NGS, together with other molecular techniques, is key to the integration of personalized medicine approaches into healthcare services. The use of NGS targeted panels, which focus on a limited and relevant set of genes or gene regions that have known associations with a particular pathology, produce large quantities of genetic information with diagnostic, prognostic and theranostic value with a high sensitivity. The simultaneous analysis of an elevated number of genes (15-200) is more resource efficient as it drastically reduces the cost and time required to obtain such genetic information enabling a more precise diagnosis and prognosis. Furthermore, the use of NGS permits the detection of emerging clones which can help inform disease follow-up and may be associated with treatment resistance, thus providing data that can help guide individualized patient therapeutic plans.

In 2016 our team implemented NGS into the routine diagnosis and prognosis of patients with acute myeloid leukemia[62]. Since then, the use of NGS has expanded to include a targeted myeloid panel for the diagnosis of patients with myeloproliferative neoplasms and myelodysplastic syndromes, a chronic lymphocytic leukemia-specific panel, and a panel for the detection of germline hematologic malignancies. However, the molecular analysis of B-cell lymphoma samples in our center is currently limited to the qPCR-based analysis of several individual genes with prognostic value (including MYD88, TP53, and EZH2) to complement the conventional cytometry, IHC and FISH tests used in routine clinical practice.

Several commercial gene panels are currently available on the market for the detection of mutations with diagnostic, prognostic or theranostic value in DLBCL and FL, given the considerable overlap of genetic alterations between GCB DLBCL and FL[32], including Oncomine™ Lymphoma (ThermoFisher), FusionPlex® Lymphoma (Archer) and Lymphoma Solution® (SOPHiA).

Incorporating a comprehensive NGS-based characterization of somatic gene mutations as a precision medicine strategy for B-cell lymphomas would assist in the daily practice by refining DLBCL and FL classification and prognosis. Importantly, it would also facilitate individualized therapeutic decision-making for patients and increase treatment opportunities by identifying candidates for clinical trials with targeted therapies. However, feasibility studies would be required to determine the clinical utility and added value of incorporating an NGS panel in the multidisciplinary diagnostic work-up, since “while many stakeholders believe that personalized medicine can provide benefits to patients and the healthcare system, payer and providers are often reluctant to change policies and practices without convincing evidence of clinical and economic value”[63].

It is also important to consider the limitations of introducing such molecular analyses for B-cell lymphomas into routine hematology laboratories. Difficulties arise in interpretation of the results generated by extensive NGS panels due to the data’s complexity and uncertainty about the biological relevance as not all molecular variants are clinically actionable. For this reason, it is essential to have highly trained staff with experience in the interpretation of the clinical impact of tumor variants. Other limitations include the economic cost of molecular analyses and the turnaround time, which has a large impact on the applicability of genomic tests to clinical decision-making. The potential to multiplex lymphoma samples with other targeted panels in the same sequencing run would help optimize the resources dedicated to library preparation and sequencing, and minimize the time required to analyze patient samples and report results to guide clinical decision-making. This is essential for aggressive B-cell lymphomas where immediate treatment is frequently required.

CONCLUSION

The incorporation of molecular testing into the routine clinical management of patients with B-cell lymphomas via the implementation of a targeted NGS panel would help improve disease subtype classification, allow the prediction of therapeutic outcome to currently available treatments, and identify patients for personalized treatment. Moreover, the optimization of non-invasive ctDNA analysis could allow for closer patient monitoring and earlier relapse detection.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Hematology

Country/Territory of origin: Spain

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Luo Y, China; Shahriari M, Iran S-Editor: Liu JH L-Editor: A P-Editor: Cai YX

References
1.  Alaggio R, Amador C, Anagnostopoulos I, Attygalle AD, Araujo IBO, Berti E, Bhagat G, Borges AM, Boyer D, Calaminici M, Chadburn A, Chan JKC, Cheuk W, Chng WJ, Choi JK, Chuang SS, Coupland SE, Czader M, Dave SS, de Jong D, Du MQ, Elenitoba-Johnson KS, Ferry J, Geyer J, Gratzinger D, Guitart J, Gujral S, Harris M, Harrison CJ, Hartmann S, Hochhaus A, Jansen PM, Karube K, Kempf W, Khoury J, Kimura H, Klapper W, Kovach AE, Kumar S, Lazar AJ, Lazzi S, Leoncini L, Leung N, Leventaki V, Li XQ, Lim MS, Liu WP, Louissaint A Jr, Marcogliese A, Medeiros LJ, Michal M, Miranda RN, Mitteldorf C, Montes-Moreno S, Morice W, Nardi V, Naresh KN, Natkunam Y, Ng SB, Oschlies I, Ott G, Parrens M, Pulitzer M, Rajkumar SV, Rawstron AC, Rech K, Rosenwald A, Said J, Sarkozy C, Sayed S, Saygin C, Schuh A, Sewell W, Siebert R, Sohani AR, Tooze R, Traverse-Glehen A, Vega F, Vergier B, Wechalekar AD, Wood B, Xerri L, Xiao W. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia. 2022;36:1720-1748.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Bastos-Oreiro M, Muntañola A, Panizo C, Gonzalez-Barca E, de Villambrosia SG, Córdoba R, López JLB, González-Sierra P, Terol MJ, Gutierrez A, Grande C, Ramirez MJ, Iserte L, Perez E, Navarro B, Gomez P, Salar A, Luzardo H, López A, Del Campo R, García-Belmonte D, Vida MJ, Infante M, Queizan-Hernandez JA, Novelli S, Moreno M, Penarrubia M, Gómez J, Domingo A, Donato E, Viguria MC, López F, Rodriguez MJ, Pardal E, Noriega V, Andreu R, Peñalver J, Martín A, Caballero D, López-Guillermo A. RELINF: prospective epidemiological registry of lymphoid neoplasms in Spain. A project from the GELTAMO group. Ann Hematol. 2020;99:799-808.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Casulo C, Byrtek M, Dawson KL, Zhou X, Farber CM, Flowers CR, Hainsworth JD, Maurer MJ, Cerhan JR, Link BK, Zelenetz AD, Friedberg JW. 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;33:2516-2522.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Hans CP, Weisenburger DD, Greiner TC, Gascoyne RD, Delabie J, Ott G, Müller-Hermelink HK, Campo E, Braziel RM, Jaffe ES, Pan Z, Farinha P, Smith LM, Falini B, Banham AH, Rosenwald A, Staudt LM, Connors JM, Armitage JO, Chan WC. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004;103:275-282.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermelink HK, Smeland EB, Giltnane JM, Hurt EM, Zhao H, Averett L, Yang L, Wilson WH, Jaffe ES, Simon R, Klausner RD, Powell J, Duffey PL, Longo DL, Greiner TC, Weisenburger DD, Sanger WG, Dave BJ, Lynch JC, Vose J, Armitage JO, Montserrat E, López-Guillermo A, Grogan TM, Miller TP, LeBlanc M, Ott G, Kvaloy S, Delabie J, Holte H, Krajci P, Stokke T, Staudt LM; Lymphoma/Leukemia Molecular Profiling Project. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med. 2002;346:1937-1947.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Friedberg JW. How I treat double-hit lymphoma. Blood. 2017;130:590-596.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Pedersen MØ, Gang AO, Poulsen TS, Knudsen H, Lauritzen AF, Nielsen SL, Klausen TW, Nørgaard P. MYC translocation partner gene determines survival of patients with large B-cell lymphoma with MYC- or double-hit MYC/BCL2 translocations. Eur J Haematol. 2014;92:42-48.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma-treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11:12-23.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Lee HJ, Shin DH, Kim KB, Shin N, Park WY, Lee JH, Choi KU, Kim JY, Lee CH, Sol MY. Polycomb protein EZH2 expression in diffuse large B-cell lymphoma is associated with better prognosis in patients treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone. Leuk Lymphoma. 2014;55:2056-2063.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Leich E, Salaverria I, Bea S, Zettl A, Wright G, Moreno V, Gascoyne RD, Chan WC, Braziel RM, Rimsza LM, Weisenburger DD, Delabie J, Jaffe ES, Lister A, Fitzgibbon J, Staudt LM, Hartmann EM, Mueller-Hermelink HK, Campo E, Ott G, Rosenwald A. Follicular lymphomas with and without translocation t(14;18) differ in gene expression profiles and genetic alterations. Blood. 2009;114:826-834.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Nann D, Ramis-Zaldivar JE, Müller I, Gonzalez-Farre B, Schmidt J, Egan C, Salmeron-Villalobos J, Clot G, Mattern S, Otto F, Mankel B, Colomer D, Balagué O, Szablewski V, Lome-Maldonado C, Leoncini L, Dojcinov S, Chott A, Copie-Bergman C, Bonzheim I, Fend F, Jaffe ES, Campo E, Salaverria I, Quintanilla-Martinez L. Follicular lymphoma t(14;18)-negative is genetically a heterogeneous disease. Blood Adv. 2020;4:5652-5665.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Solal-Céligny P, Roy P, Colombat P, White J, Armitage JO, Arranz-Saez R, Au WY, Bellei M, Brice P, Caballero D, Coiffier B, Conde-Garcia E, Doyen C, Federico M, Fisher RI, Garcia-Conde JF, Guglielmi C, Hagenbeek A, Haïoun C, LeBlanc M, Lister AT, Lopez-Guillermo A, McLaughlin P, Milpied N, Morel P, Mounier N, Proctor SJ, Rohatiner A, Smith P, Soubeyran P, Tilly H, Vitolo U, Zinzani PL, Zucca E, Montserrat E. Follicular lymphoma international prognostic index. Blood. 2004;104:1258-1265.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Federico M, Bellei M, Marcheselli L, Luminari S, Lopez-Guillermo A, Vitolo U, Pro B, Pileri S, Pulsoni A, Soubeyran P, Cortelazzo S, Martinelli G, Martelli M, Rigacci L, Arcaini L, Di Raimondo F, Merli F, Sabattini E, McLaughlin P, Solal-Céligny P. 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. 2009;27:4555-4562.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Leonard JP. POD24 in follicular lymphoma: time to be "wise". Blood. 2022;139:1609-1610.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Dreyling M, Ghielmini M, Rule S, Salles G, Vitolo U, Ladetto M; ESMO Guidelines Committee. Newly diagnosed and relapsed follicular lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27:v83-v90.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Campo E, Jaffe ES, Cook JR, Quintanilla-Martinez L, Swerdlow SH, Anderson KC, Brousset P, Cerroni L, de Leval L, Dirnhofer S, Dogan A, Feldman AL, Fend F, Friedberg JW, Gaulard P, Ghia P, Horwitz SM, King RL, Salles G, San-Miguel J, Seymour JF, Treon SP, Vose JM, Zucca E, Advani R, Ansell S, Au WY, Barrionuevo C, Bergsagel L, Chan WC, Cohen JI, d'Amore F, Davies A, Falini B, Ghobrial IM, Goodlad JR, Gribben JG, Hsi ED, Kahl BS, Kim WS, Kumar S, LaCasce AS, Laurent C, Lenz G, Leonard JP, Link MP, Lopez-Guillermo A, Mateos MV, Macintyre E, Melnick AM, Morschhauser F, Nakamura S, Narbaitz M, Pavlovsky A, Pileri SA, Piris M, Pro B, Rajkumar V, Rosen ST, Sander B, Sehn L, Shipp MA, Smith SM, Staudt LM, Thieblemont C, Tousseyn T, Wilson WH, Yoshino T, Zinzani PL, Dreyling M, Scott DW, Winter JN, Zelenetz AD. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood. 2022;140:1229-1253.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Duncavage EJ, Bagg A, Hasserjian RP, DiNardo CD, Godley LA, Iacobucci I, Jaiswal S, Malcovati L, Vannucchi AM, Patel KP, Arber DA, Arcila ME, Bejar R, Berliner N, Borowitz MJ, Branford S, Brown AL, Cargo CA, Döhner H, Falini B, Garcia-Manero G, Haferlach T, Hellström-Lindberg E, Kim AS, Klco JM, Komrokji R, Lee-Cheun Loh M, Loghavi S, Mullighan CG, Ogawa S, Orazi A, Papaemmanuil E, Reiter A, Ross DM, Savona M, Shimamura A, Skoda RC, Solé F, Stone RM, Tefferi A, Walter MJ, Wu D, Ebert BL, Cazzola M. Genomic profiling for clinical decision making in myeloid neoplasms and acute leukemia. Blood. 2022;140:2228-2247.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  de Leval L, Alizadeh AA, Bergsagel PL, Campo E, Davies A, Dogan A, Fitzgibbon J, Horwitz SM, Melnick AM, Morice WG, Morin RD, Nadel B, Pileri SA, Rosenquist R, Rossi D, Salaverria I, Steidl C, Treon SP, Zelenetz AD, Advani RH, Allen CE, Ansell SM, Chan WC, Cook JR, Cook LB, d'Amore F, Dirnhofer S, Dreyling M, Dunleavy K, Feldman AL, Fend F, Gaulard P, Ghia P, Gribben JG, Hermine O, Hodson DJ, Hsi ED, Inghirami G, Jaffe ES, Karube K, Kataoka K, Klapper W, Kim WS, King RL, Ko YH, LaCasce AS, Lenz G, Martin-Subero JI, Piris MA, Pittaluga S, Pasqualucci L, Quintanilla-Martinez L, Rodig SJ, Rosenwald A, Salles GA, San-Miguel J, Savage KJ, Sehn LH, Semenzato G, Staudt LM, Swerdlow SH, Tam CS, Trotman J, Vose JM, Weigert O, Wilson WH, Winter JN, Wu CJ, Zinzani PL, Zucca E, Bagg A, Scott DW. Genomic profiling for clinical decision making in lymphoid neoplasms. Blood. 2022;140:2193-2227.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Rosenquist R, Beà S, Du MQ, Nadel B, Pan-Hammarström Q. Genetic landscape and deregulated pathways in B-cell lymphoid malignancies. J Intern Med. 2017;282:371-394.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Bohers E, Mareschal S, Bouzelfen A, Marchand V, Ruminy P, Maingonnat C, Ménard AL, Etancelin P, Bertrand P, Dubois S, Alcantara M, Bastard C, Tilly H, Jardin F. Targetable activating mutations are very frequent in GCB and ABC diffuse large B-cell lymphoma. Genes Chromosomes Cancer. 2014;53:144-153.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Ngo VN, Young RM, Schmitz R, Jhavar S, Xiao W, Lim KH, Kohlhammer H, Xu W, Yang Y, Zhao H, Shaffer AL, Romesser P, Wright G, Powell J, Rosenwald A, Muller-Hermelink HK, Ott G, Gascoyne RD, Connors JM, Rimsza LM, Campo E, Jaffe ES, Delabie J, Smeland EB, Fisher RI, Braziel RM, Tubbs RR, Cook JR, Weisenburger DD, Chan WC, Staudt LM. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011;470:115-119.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Mottok A, Wright G, Rosenwald A, Ott G, Ramsower C, Campo E, Braziel RM, Delabie J, Weisenburger DD, Song JY, Chan WC, Cook JR, Fu K, Greiner T, Smeland E, Holte H, Savage KJ, Glinsmann-Gibson BJ, Gascoyne RD, Staudt LM, Jaffe ES, Connors JM, Scott DW, Steidl C, Rimsza LM. Molecular classification of primary mediastinal large B-cell lymphoma using routinely available tissue specimens. Blood. 2018;132:2401-2405.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Jiang Y, Redmond D, Nie K, Eng KW, Clozel T, Martin P, Tan LH, Melnick AM, Tam W, Elemento O. Deep sequencing reveals clonal evolution patterns and mutation events associated with relapse in B-cell lymphomas. Genome Biol. 2014;15:432.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Hilton LK, Tang J, Ben-Neriah S, Alcaide M, Jiang A, Grande BM, Rushton CK, Boyle M, Meissner B, Scott DW, Morin RD. The double-hit signature identifies double-hit diffuse large B-cell lymphoma with genetic events cryptic to FISH. Blood. 2019;134:1528-1532.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, Lih CJ, Williams PM, Shaffer AL, Gerecitano J, de Vos S, Goy A, Kenkre VP, Barr PM, Blum KA, Shustov A, Advani R, Fowler NH, Vose JM, Elstrom RL, Habermann TM, Barrientos JC, McGreivy J, Fardis M, Chang BY, Clow F, Munneke B, Moussa D, Beaupre DM, Staudt LM. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922-926.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Reddy A, Zhang J, Davis NS, Moffitt AB, Love CL, Waldrop A, Leppa S, Pasanen A, Meriranta L, Karjalainen-Lindsberg ML, Nørgaard P, Pedersen M, Gang AO, Høgdall E, Heavican TB, Lone W, Iqbal J, Qin Q, Li G, Kim SY, Healy J, Richards KL, Fedoriw Y, Bernal-Mizrachi L, Koff JL, Staton AD, Flowers CR, Paltiel O, Goldschmidt N, Calaminici M, Clear A, Gribben J, Nguyen E, Czader MB, Ondrejka SL, Collie A, Hsi ED, Tse E, Au-Yeung RKH, Kwong YL, Srivastava G, Choi WWL, Evens AM, Pilichowska M, Sengar M, Reddy N, Li S, Chadburn A, Gordon LI, Jaffe ES, Levy S, Rempel R, Tzeng T, Happ LE, Dave T, Rajagopalan D, Datta J, Dunson DB, Dave SS. Genetic and Functional Drivers of Diffuse Large B Cell Lymphoma. Cell. 2017;171:481-494.e15.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Schmitz R, Wright GW, Huang DW, Johnson CA, Phelan JD, Wang JQ, Roulland S, Kasbekar M, Young RM, Shaffer AL, Hodson DJ, Xiao W, Yu X, Yang Y, Zhao H, Xu W, Liu X, Zhou B, Du W, Chan WC, Jaffe ES, Gascoyne RD, Connors JM, Campo E, Lopez-Guillermo A, Rosenwald A, Ott G, Delabie J, Rimsza LM, Tay Kuang Wei K, Zelenetz AD, Leonard JP, Bartlett NL, Tran B, Shetty J, Zhao Y, Soppet DR, Pittaluga S, Wilson WH, Staudt LM. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N Engl J Med. 2018;378:1396-1407.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Wilson WH, Wright GW, Huang DW, Hodkinson B, Balasubramanian S, Fan Y, Vermeulen J, Shreeve M, Staudt LM. Effect of ibrutinib with R-CHOP chemotherapy in genetic subtypes of DLBCL. Cancer Cell. 2021;39:1643-1653.e3.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Chapuy B, Stewart C, Dunford AJ, Kim J, Kamburov A, Redd RA, Lawrence MS, Roemer MGM, Li AJ, Ziepert M, Staiger AM, Wala JA, Ducar MD, Leshchiner I, Rheinbay E, Taylor-Weiner A, Coughlin CA, Hess JM, Pedamallu CS, Livitz D, Rosebrock D, Rosenberg M, Tracy AA, Horn H, van Hummelen P, Feldman AL, Link BK, Novak AJ, Cerhan JR, Habermann TM, Siebert R, Rosenwald A, Thorner AR, Meyerson ML, Golub TR, Beroukhim R, Wulf GG, Ott G, Rodig SJ, Monti S, Neuberg DS, Loeffler M, Pfreundschuh M, Trümper L, Getz G, Shipp MA. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24:679-690.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Karube K, Enjuanes A, Dlouhy I, Jares P, Martin-Garcia D, Nadeu F, Ordóñez GR, Rovira J, Clot G, Royo C, Navarro A, Gonzalez-Farre B, Vaghefi A, Castellano G, Rubio-Perez C, Tamborero D, Briones J, Salar A, Sancho JM, Mercadal S, Gonzalez-Barca E, Escoda L, Miyoshi H, Ohshima K, Miyawaki K, Kato K, Akashi K, Mozos A, Colomo L, Alcoceba M, Valera A, Carrió A, Costa D, Lopez-Bigas N, Schmitz R, Staudt LM, Salaverria I, López-Guillermo A, Campo E. Integrating genomic alterations in diffuse large B-cell lymphoma identifies new relevant pathways and potential therapeutic targets. Leukemia. 2018;32:675-684.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Pasqualucci L, Dominguez-Sola D, Chiarenza A, Fabbri G, Grunn A, Trifonov V, Kasper LH, Lerach S, Tang H, Ma J, Rossi D, Chadburn A, Murty VV, Mullighan CG, Gaidano G, Rabadan R, Brindle PK, Dalla-Favera R. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature. 2011;471:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Okosun J, Bödör C, Wang J, Araf S, Yang CY, Pan C, Boller S, Cittaro D, Bozek M, Iqbal S, Matthews J, Wrench D, Marzec J, Tawana K, Popov N, O'Riain C, O'Shea D, Carlotti E, Davies A, Lawrie CH, Matolcsy A, Calaminici M, Norton A, Byers RJ, Mein C, Stupka E, Lister TA, Lenz G, Montoto S, Gribben JG, Fan Y, Grosschedl R, Chelala C, Fitzgibbon J. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat Genet. 2014;46:176-181.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Pasqualucci L, Khiabanian H, Fangazio M, Vasishtha M, Messina M, Holmes AB, Ouillette P, Trifonov V, Rossi D, Tabbò F, Ponzoni M, Chadburn A, Murty VV, Bhagat G, Gaidano G, Inghirami G, Malek SN, Rabadan R, Dalla-Favera R. Genetics of follicular lymphoma transformation. Cell Rep. 2014;6:130-140.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sander CA, Yano T, Clark HM, Harris C, Longo DL, Jaffe ES, Raffeld M. p53 mutation is associated with progression in follicular lymphomas. Blood. 1993;82:1994-2004.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  O'Shea D, O'Riain C, Taylor C, Waters R, Carlotti E, Macdougall F, Gribben J, Rosenwald A, Ott G, Rimsza LM, Smeland EB, Johnson N, Campo E, Greiner TC, Chan WC, Gascoyne RD, Wright G, Staudt LM, Lister TA, Fitzgibbon J. 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;112:3126-3129.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Cheung KJ, Shah SP, Steidl C, Johnson N, Relander T, Telenius A, Lai B, Murphy KP, Lam W, Al-Tourah AJ, Connors JM, Ng RT, Gascoyne RD, Horsman DE. Genome-wide profiling of follicular lymphoma by array comparative genomic hybridization reveals prognostically significant DNA copy number imbalances. Blood. 2009;113:137-148.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Cahill KE, Smith SM. Follicular Lymphoma: a Focus on Current and Emerging Therapies. Oncology (Williston Park). 2022;36:97-106.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Pastore A, Jurinovic V, Kridel R, Hoster E, Staiger AM, Szczepanowski M, Pott C, Kopp N, Murakami M, Horn H, Leich E, Moccia AA, Mottok A, Sunkavalli A, Van Hummelen P, Ducar M, Ennishi D, Shulha HP, Hother C, Connors JM, Sehn LH, Dreyling M, Neuberg D, Möller P, Feller AC, Hansmann ML, Stein H, Rosenwald A, Ott G, Klapper W, Unterhalt M, Hiddemann W, Gascoyne RD, Weinstock DM, Weigert O. 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;16:1111-1122.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Martínez-Laperche C, Sanz-Villanueva L, Díaz Crespo FJ, Muñiz P, Martín Rojas R, Carbonell D, Chicano M, Suárez-González J, Menárguez J, Kwon M, Diez Martín JL, Buño I, Bastos Oreiro M. EZH2 mutations at diagnosis in follicular lymphoma: a promising biomarker to guide frontline treatment. BMC Cancer. 2022;22:982.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Lockmer S, Ren W, Brodtkorb M, Østenstad B, Wahlin BE, Pan-Hammarström Q, Kimby E. M7-FLIPI is not prognostic in follicular lymphoma patients with first-line rituximab chemo-free therapy. Br J Haematol. 2020;188:259-267.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Cartron G, Trotman J. Time for an individualized approach to first-line management of follicular lymphoma. Haematologica. 2022;107:7-18.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Kalakonda N, Maerevoet M, Cavallo F, Follows G, Goy A, Vermaat JSP, Casasnovas O, Hamad N, Zijlstra JM, Bakhshi S, Bouabdallah R, Choquet S, Gurion R, Hill B, Jaeger U, Sancho JM, Schuster M, Thieblemont C, De la Cruz F, Egyed M, Mishra S, Offner F, Vassilakopoulos TP, Warzocha K, McCarthy D, Ma X, Corona K, Saint-Martin JR, Chang H, Landesman Y, Joshi A, Wang H, Shah J, Shacham S, Kauffman M, Van Den Neste E, Canales MA. Selinexor in patients with relapsed or refractory diffuse large B-cell lymphoma (SADAL): a single-arm, multinational, multicentre, open-label, phase 2 trial. Lancet Haematol. 2020;7:e511-e522.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Sehn LH, Herrera AF, Flowers CR, Kamdar MK, McMillan A, Hertzberg M, Assouline S, Kim TM, Kim WS, Ozcan M, Hirata J, Penuel E, Paulson JN, Cheng J, Ku G, Matasar MJ. Polatuzumab Vedotin in Relapsed or Refractory Diffuse Large B-Cell Lymphoma. J Clin Oncol. 2020;38:155-165.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Morschhauser F, Tilly H, Chaidos A, McKay P, Phillips T, Assouline S, Batlevi CL, Campbell P, Ribrag V, Damaj GL, Dickinson M, Jurczak W, Kazmierczak M, Opat S, Radford J, Schmitt A, Yang J, Whalen J, Agarwal S, Adib D, Salles G. Tazemetostat for patients with relapsed or refractory follicular lymphoma: an open-label, single-arm, multicentre, phase 2 trial. Lancet Oncol. 2020;21:1433-1442.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Gopal AK, Kahl BS, de Vos S, Wagner-Johnston ND, Schuster SJ, Jurczak WJ, Flinn IW, Flowers CR, Martin P, Viardot A, Blum KA, Goy AH, Davies AJ, Zinzani PL, Dreyling M, Johnson D, Miller LL, Holes L, Li D, Dansey RD, Godfrey WR, Salles GA. PI3Kδ inhibition by idelalisib in patients with relapsed indolent lymphoma. N Engl J Med. 2014;370:1008-1018.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Dreyling M, Santoro A, Mollica L, Leppä S, Follows GA, Lenz G, Kim WS, Nagler A, Panayiotidis P, Demeter J, Özcan M, Kosinova M, Bouabdallah K, Morschhauser F, Stevens DA, Trevarthen D, Giurescu M, Cupit L, Liu L, Köchert K, Seidel H, Peña C, Yin S, Hiemeyer F, Garcia-Vargas J, Childs BH, Zinzani PL. Phosphatidylinositol 3-Kinase Inhibition by Copanlisib in Relapsed or Refractory Indolent Lymphoma. J Clin Oncol. 2017;35:3898-3905.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Flinn IW, Miller CB, Ardeshna KM, Tetreault S, Assouline SE, Mayer J, Merli M, Lunin SD, Pettitt AR, Nagy Z, Tournilhac O, Abou-Nassar KE, Crump M, Jacobsen ED, de Vos S, Kelly VM, Shi W, Steelman L, Le N, Weaver DT, Lustgarten S, Wagner-Johnston ND, Zinzani PL. DYNAMO: A Phase II Study of Duvelisib (IPI-145) in Patients With Refractory Indolent Non-Hodgkin Lymphoma. J Clin Oncol. 2019;37:912-922.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Fowler NH, Samaniego F, Jurczak W, Ghosh N, Derenzini E, Reeves JA, Knopińska-Posłuszny W, Cheah CY, Phillips T, Lech-Maranda E, Cheson BD, Caimi PF, Grosicki S, Leslie LA, Chavez JC, Fonseca G, Babu S, Hodson DJ, Shao SH, Burke JM, Sharman JP, Law JY, Pagel JM, Miskin HP, Sportelli P, O'Connor OA, Weiss MS, Zinzani PL. Umbralisib, a Dual PI3Kδ/CK1ε Inhibitor in Patients With Relapsed or Refractory Indolent Lymphoma. J Clin Oncol. 2021;39:1609-1618.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Ramos CA, Heslop HE, Brenner MK. CAR-T Cell Therapy for Lymphoma. Annu Rev Med. 2016;67:165-183.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest. 2015;125:3392-3400.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, Mehta A, Purev E, Maloney DG, Andreadis C, Sehgal A, Solomon SR, Ghosh N, Albertson TM, Garcia J, Kostic A, Mallaney M, Ogasawara K, Newhall K, Kim Y, Li D, Siddiqi T. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396:839-852.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Fowler NH, Dickinson M, Dreyling M, Martinez-Lopez J, Kolstad A, Butler J, Ghosh M, Popplewell L, Chavez JC, Bachy E, Kato K, Harigae H, Kersten MJ, Andreadis C, Riedell PA, Ho PJ, Pérez-Simón JA, Chen AI, Nastoupil LJ, von Tresckow B, Ferreri AJM, Teshima T, Patten PEM, McGuirk JP, Petzer AL, Offner F, Viardot A, Zinzani PL, Malladi R, Zia A, Awasthi R, Masood A, Anak O, Schuster SJ, Thieblemont C. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med. 2022;28:325-332.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Locke FL, Miklos DB, Jacobson CA, Perales MA, Kersten MJ, Oluwole OO, Ghobadi A, Rapoport AP, McGuirk J, Pagel JM, Muñoz J, Farooq U, van Meerten T, Reagan PM, Sureda A, Flinn IW, Vandenberghe P, Song KW, Dickinson M, Minnema MC, Riedell PA, Leslie LA, Chaganti S, Yang Y, Filosto S, Shah J, Schupp M, To C, Cheng P, Gordon LI, Westin JR; All ZUMA-7 Investigators and Contributing Kite Members. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N Engl J Med. 2022;386:640-654.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Roschewski M, Dunleavy K, Pittaluga S, Moorhead M, Pepin F, Kong K, Shovlin M, Jaffe ES, Staudt LM, Lai C, Steinberg SM, Chen CC, Zheng J, Willis TD, Faham M, Wilson WH. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16:541-549.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Wang H, Du X, Chen WH, Lou J, Xiao HL, Pan YM, Chen H, An N, Zhang QX. Establishment of a Quantitative Polymerase Chain Reaction Assay for Monitoring Chimeric Antigen Receptor T Cells in Peripheral Blood. Transplant Proc. 2018;50:104-109.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Gonçalves MC, de Oliveira CRGCM, Sandes AF, Rodrigues CA, Novis Y, Viana PCC, Serra MMP, Zerbini MCN. Core Needle Biopsy in Lymphoma Diagnosis: The Diagnostic Performance and the Role of the Multidisciplinary Approach in the Optimization of Results. Am J Surg Pathol. 2023;47:111-123.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Kurtz DM, Scherer F, Jin MC, Soo J, Craig AFM, Esfahani MS, Chabon JJ, Stehr H, Liu CL, Tibshirani R, Maeda LS, Gupta NK, Khodadoust MS, Advani RH, Levy R, Newman AM, Dührsen U, Hüttmann A, Meignan M, Casasnovas RO, Westin JR, Roschewski M, Wilson WH, Gaidano G, Rossi D, Diehn M, Alizadeh AA. Circulating Tumor DNA Measurements As Early Outcome Predictors in Diffuse Large B-Cell Lymphoma. J Clin Oncol. 2018;36:2845-2853.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Gormally E, Caboux E, Vineis P, Hainaut P. Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res. 2007;635:105-117.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Scherer F, Kurtz DM, Newman AM, Stehr H, Craig AF, Esfahani MS, Lovejoy AF, Chabon JJ, Klass DM, Liu CL, Zhou L, Glover C, Visser BC, Poultsides GA, Advani RH, Maeda LS, Gupta NK, Levy R, Ohgami RS, Kunder CA, Diehn M, Alizadeh AA. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8:364ra155.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Heitzer E, Haque IS, Roberts CES, Speicher MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet. 2019;20:71-88.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines)  Non-small cell lung cancer. Version 6.2020. Published 2017. Accessed July 20, 2020. Available from: http://www.nccn.org.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Stuckey R, Bilbao-Sieyro C, Gómez-Casares MT. A summary of the molecular testing recommended in acute myeloid leukemia. Ann Mol Genet Med. 2020;4:012-017.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Novartis Oncology  The precision oncology annual trend report: perspectives from payers, oncologists, and pathologists. Published 2016. Accessed July 21, 2020. Available from: http://www.hcp.novartis.com/care-management.  [PubMed]  [DOI]  [Cited in This Article: ]