Plasmablastic Lymphoma. A State-of-the-Art Review: Part 2-Focus on Therapy  

Michele Bibas.

Department of Clinical Research, Hematology. National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.S.S. Via Portuense 292 00148 Rome Italy.

Correspondence to: Michele Bibas. Department of Clinical Research, Hematology. National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.S.S. Via Portuense 292 00148 Rome Italy. E-mail: Michele.bibas@inmi.it

Published: March 01, 2024
Received: January 25, 2024
Accepted: February 14, 2024
Mediterr J Hematol Infect Dis 2024, 16(1): e2024015 DOI 10.4084/MJHID.2024.015

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The objective of this two-part review is to present a current and comprehensive understanding of the diagnosis and management of plasmablastic lymphoma. The first part, which was published previously, focused on the study of epidemiology, etiology, clinicopathological characteristics, differential diagnosis, prognostic variables, and the impact of plasmablastic lymphoma on specific populations. This second part addresses the difficult topic of the treatment of plasmablastic lymphoma, specifically examining both the conventional, consolidated approach and the novel therapeutic strategy.



Article Highlights


Introduction

The identification and characterization of plasmablastic lymphoma (PBL) date originally from 1992,[1] after that, it was recognized as a distinct form of oral lymphoma occurring in individuals who are HIV-positive.[2] This particular subtype of large B-cell lymphoma (LBCL) is now categorized as a distinct entity, with extranodal disease being the prevailing characteristic.[3-6]
Despite earlier studies indicating a strong association, it has been observed that around 50% to 60% of cases are associated with HIV. 3–5 PBL accounts for just 2% of lymphomas in individuals who are HIV-positive.[3-6] The disease has the potential to impact individuals with compromised immune systems as well as those with robust immune systems.
Histological examination reveals the presence of several neoplastic plasmablasts and immunoblasts. The identified cells exhibit a significant proliferation index and possess a plasma cell immunophenotype, characterized by the presence of plasma cell markers and a limited or missing expression of B cell markers.
Notably, transcription factors linked to plasmacytic differentiation, including CD38, CD138, MUM1, Blimp1, and XBP1, are present. The absence of CD20 and PAX5 has been very frequently observed. PBL frequently has a connection to the overexpression of the MYC gene, which can result from translocations, amplifications, or constitutive STAT3 activation. This is in contrast to plasma cell neoplasms.[3-7] Moreover, around 80% of PBL cases were found to have a simultaneous Epstein-Barr virus (EBV) infection. This feature can be regarded as a discerning element differentiating PBL from plasmablastic myeloma. Hence, distinguishing between plasmablastic myeloma, lymphomas, and PBL can pose a significant difficulty. Translational research encounters several challenges, such as the limited occurrence of PBL and the absence of an established treatment strategy due to a scarcity of thorough clinical data.[3-8]
Historically, PBL has been associated with a negative outlook, as early estimates of median overall survival (OS) ranged from 8 to 15 months.[3-6] There have been more survival estimates reported in recent literature that display significant diversity.
A population-based SEER survival analysis examined 248 patients who had treatment between 2010 and 2016. The analysis revealed that the median overall survival was 47 months.[9]
Further, a total of 1,800 patients were very recently assessed using SEER and the NCDB. An exceptional median overall survival of 58.6 months was seen in the treated patients.[10]
Although there have been some improvements recently, CD38-directed monoclonal antibody therapy, proteasome inhibitors, and immunomodulatory therapeutic regimens, along with intensive polychemotherapy, are still rarely available for older and weaker patients. Those limitations highlight the requirement for therapeutic strategies that are precisely personalized to fulfill individual requirements.


Treatment

Establishing a therapeutic standard is quite problematic due to the rarity of the disease and the lack of controlled trials available for comparing different treatments. In this second part of the review, our intent is to provide a comprehensive and detailed description of the achievements of the diverse consolidated therapies, as well as those that are currently being developed. We will organize the information into pertinent chapters for clarity.

Standard Polychemotherapy


Patients with untreated PBL exhibit a median overall survival of 3 months in individuals who are HIV-positive and 4 months in those who are HIV-negative.[3-6]
Patients diagnosed with PBL have been subjected to a wide range of treatment options, covering localized disease management by radiation as well as the administration of diverse chemotherapy combinations. Individuals who are diagnosed with limited-stage disease tend to have a more favorable prognosis, and in certain instances, aggressive treatment measures may not be recommended. Disease control may be achieved through the utilization of a combination therapy involving doxorubicin-based chemotherapy and radiation therapy.[3-6] Of note, the majority of PBL patients must be considered high-risk patients and treated with polychemotherapy. Polychemotherapy has yielded complete remissions (CRs) in nearly 50% of patients with disseminated disease. The response and relapse rates among different first-line regimens are presented in Table 1.


Table 1
Table 1. Literature review of the polychemotherapy regimens of PBL patients.

However, a significant proportion of patients, approximately 70%, inevitably die of progressive disease. At present, there is a lack of established care standards that delineate the most effective therapy method. Throughout history, the use of CHOP has been widely adopted as the primary therapeutic approach for PBL, with a particular emphasis on its utilization in nations with lower economic resources. According to the latest National Comprehensive Cancer Network (NCCN) guidelines on B-cell lymphomas (version 1.2024, January 18, 2024),[22] it has been determined that CHOP is not adequate as a first-line therapy.
NCCN supports using more intensive treatment plans for PBL and suggests dose-adjusted (DA)-EPOCH as an alternative way to treat the disease. DA-EPOCH includes etoposide, vincristine, doxorubicin, cyclophosphamide, and prednisone given in bolus doses. Some other treatment plans that have been suggested are CODOX-M/IVAC (modified) and HyperCVAD (which includes cyclophosphamide, vincristine, doxorubicin, dexamethasone, and high doses of methotrexate and cytarabine). Furthermore, these guidelines suggest the potential use of high-dose therapy with autologous stem cell rescue during the first complete remission for a particular subgroup of patients at high risk. A high-risk factor includes an International Prognostic Index (IPI) score above 2 and changes to the MYC gene or deletion of the TP53 gene. It is important to acknowledge that individuals who are HIV-negative and diagnosed with plasmablastic lymphoma are commonly recognized as having a condition associated with an elevated risk. Typically, those who are HIV-negative and diagnosed with plasmablastic lymphoma are generally characterized as having a more threatening disease. On the other hand, there is a more favorable outlook for individuals who are HIV-positive and diagnosed with PBL when they successfully attain complete remission with the administration of chemotherapy. Antiretroviral therapy is crucial for improving the management of PBL HIV+, and the attainment of complete remission (CR) has been shown to enhance the short-term prognosis of PBL.[3-6]

Bortezomib

Bortezomib is currently approved for treating adult patients with multiple myeloma and adult patients with mantle cell lymphoma. The small molecule bortezomib is a reversible proteasome inhibitor that works on the 26S proteasomes. It stops many signaling pathways by targeting a single molecular target, the proteasome. Bortezomib's anti-neoplastic effect likely involves several distinct mechanisms, such as inhibition of cell growth and survival pathways, induction of apoptosis, and inhibition of the expression of genes that regulate cellular adhesion, migration, and angiogenesis. Thus, the mechanisms by which bortezomib elicits its anti-tumor activity may differ between tumor types, as could the importance of each affected pathway in inhibiting tumor growth. Bortezomib is thought to work against multiple myeloma by stopping NF-B from working and stopping the breakdown of phosphorylated IB. Because of this, it seems like a good way to treat PBL patients, whose biological and phenotypic traits are somewhere between those of ABC-DLBCL and MM. Several reports have reported its activity in lymphoma, specifically in non-Germinal Center B-cell lymphomas such as DLBCL and mantle cell lymphoma.[23-26] Thus, bortezomib is one of the most frequently used drugs in the treatment of PBL. It has been utilized as a single agent and in conjunction with chemotherapy. The objective of our extensive literature review was to determine if patients with PBL would experience more benefits from the inclusion of bortezomib in their polychemotherapy (Table 2).

Table 2
Table 2. Literature review of Bortezomib containing regimens in PBL.

Five out of six patients treated with bortezomib alone in a small series achieved a partial response—two as first-line therapy and three as salvage therapy.[40] Bortezomib has been used in combination with CHOP as frontline therapy in three HIV-associated PBL patients, all of whom achieved a CR, and two of whom are still alive 14 and 22 months after completing V-CHOP, respectively.[37] According to a positron emission tomography scan, Castillo et al. reported three patients without relapse at 12, 18, and 24 months.[36] Recently, Dittus and Castillo reported 8 and 16 patients, respectively, with CR rates of 87.5% and 94%.[41,42] In the latter series, two patients received an ASCT for consolidation. Castillo reported a 5-year OS of 63%, while Dittus reported a 2-year PFS and OS of 50%.[41,42] Bortezomib has also been used with THP-COP (pirarubicin, cyclophosphamide, vincristine, and prednisone), ESHAP (etoposide, high-dose prednisolone, high-dose cytarabine, and platinum), ICE (ifosfamide, carboplatin, and etoposide), bendamustine, rituximab, and DT.[35] A recent systematic review found 21 patients with PBL, 11 of whom had HIV and 10 of whom did not. Eleven of them were given bortezomib as the first line of treatment, and the other 10 were given it after a relapse, either by itself or with other standard cytostatic drugs. The ORR for bortezomib-containing regimens was 100% in the frontline setting and 90% in the relapse setting. In addition, the 2-year overall survival rate for patients who received initial treatment was 55%, while the median OS for relapsed patients was 14 months.[40]
Finally, upon evaluating the existing literature, it was found that patients with PBL benefitted from including bortezomib in their treatment. However, we acknowledge that we are currently experiencing a shortage of randomized studies comparing chemotherapy regimens with and without bortezomib.


Lenalidomide

Lenalidomide is an oral immunomodulator with direct antitumor activity and immunologic effects, such as stopping tumor cell growth and angiogenesis and increasing the killing power of T- and natural killer (NK) cells in lab experiments. In vitro and in vivo studies revealed antitumor, antiproliferative, and increased NK cell number and activity against B-cell malignant lymphoma in general and against DLBCL, FL, and MCL cells. In preclinical models of activated B-cell (ABC)-subtype DLBCL, lenalidomide-induced cytotoxicity required the presence of cereblon to downregulate interferon regulatory factor-4 and B-cell receptor-NFB and boost interferon production. Of note, low cereblon expression is a potential lenalidomide resistance mechanism.[47]
According to a thorough review of the literature, lenalidomide has only been used in a few cases of PBL, and each case is very different from the others (Table 3). Cases of refractory PBL treated with lenalidomide as a single agent were reported to have a favorable response, albeit a brief one.


Table 3
Table 3. Literature review of Lenalidomide containing regimens in PBL.

It also demonstrated efficacy when combined with CHOP, or cyclophosphamide-dexamethasone.[49] Ando et al. utilized bortezomib to treat chemotherapy-resistant PBL patients, which resulted in a clinical response but was discontinued due to peripheral neuropathy.[43] The patient was then treated with a combination of lenalidomide and dexamethasone for more than two years, with a partial response that persisted.[43]
A person with PBL who was not responding to mini-CHOP as a first-line treatment went into complete remission after taking tislelizumab, a checkpoint inhibitor, and lenalidomide.[52]
Another patient with relapsed PBL with parotid involvement was treated with a combination of lenalidomide and bortezomib.[51] This patient received only two cycles of the protocol before it was discontinued due to bortezomib-induced pancreatitis. However, a PET CT scan performed after the two cycles revealed no evidence of disease, and the patient remained in complete remission for at least a year following the initiation of salvage therapy.

Brentuximab Vedotin (BV)

CD30 is a 120-kilodalton transmembrane cytokine receptor, part of the tumor necrosis factor receptor family 4. It is found on the lymphoid cells of almost all HL and ALCL patients. Expression of CD30 is restricted to activated lymphocytes and eosinophils, typically found in lymphoid tissues but not in peripheral blood cells. Thus, CD30 has been identified as a desirable therapeutic target. Brentuximab vedotin (BV) is a chimeric IgG1 anti-CD30 antibody-drug conjugated by a protease-cleavable linker to the microtubule-disrupting agent monomethyl auristatin E, which has demonstrated significant anti-tumor activity in both HL and ALCL.[54,55]
The main way that BV works is by delivering monomethyl auristatin E to tumor cells that express CD30. In addition to antibody-dependent cellular phagocytosis, immunogenic cell death, and the bystander effect, other mechanisms of tumor cell death may contribute to the clinical activity of this drug. The availability of BV has become a significant advance in the treatment of patients with relapsed and resistant HL. In addition, the significant clinical activity observed and the good tolerability of BV have allowed for widespread investigation and use of BV in a variety of lymphoma patients, and several groups are testing BV-based therapies in the management of newly diagnosed patients with HL and ALCL, with promising preliminary results.[54,55] Positive expression of CD30 has been detected in 30–50% of PBL cases, making CD30 a viable target for PBL.[3-10] It is reported that the use of brentuximab vedotin resulted in an impressive reduction in tumor size but also a fatal outcome due to tumor lysis syndrome and comorbidities.[56] The patient's tumor, which had undergone extensive prior treatment and was unresponsive to numerous chemotherapy regimens, had a positive response when treated with brentuximab vedotin as a standalone therapy and ionizing radiation. This information shows that more research should be done on brentuximab vedotin for CD30-positive PBL, either as a single treatment or in combination with standard chemotherapy.

Selinexor

The overexpression of XPO1 (exportin 1), one of eight nucleocytoplasmic shuttling proteins that help move proteins from the nucleus to the cytoplasm, is linked to a poor prognosis in DLBCL.[57] XPO1 mediates the functional inactivation of multiple tumor suppressor proteins (such as p53, p73, IkB, and FOXO) and facilitates the increased translation of oncoproteins relevant to B-cell biology and DLBCL.[58,59] By forcing these proteins to stay in the nucleus, blocking XPO1 in DLBCL may restore the tumor-suppressing and growth-regulating effects of several tumor-suppressor proteins. This may also reverse chemotherapy resistance.[60] Selinexor, an oral selective inhibitor of XPO1-mediated nuclear export, induces the expected nuclear accumulation and activation of tumor suppressor proteins and decreases the levels of Bcl2, Bcl-XL, and c-Myc oncoproteins. Based on the safety and effectiveness data from the STORM study, the US Food and Drug Administration approved the use of low-dose dexamethasone and selinexor (80 mg twice weekly) together for people with advanced refractory multiple myeloma.[61] In a phase 1 study that showed selinexor's preliminary activity in several types of blood cancer, such as myeloma and DLBCL, the single drug selinexor showed an overall response rate (ORR) of 32% in 13 of 41 patients who had already received a lot of treatment for DLBCL, and a complete response rate of 10% in 4 of those patients. Based on that study, the recommended dose was 35 mg/m2 (60 mg) twice weekly.[62] The FDA has approved selinexor to treat diffuse large B-cell lymphoma (RR DLBCL) and relapsed or refractory multiple myeloma (RR MM). It is very effective as a type of treatment.[63,64] Regarding PBL, a case of a profound response to selinexor in HIV-negative, EBV-negative, heavily pretreated young PBL patients has been reported recently. 60 mg of Selinexor were administered on days 1, 8, and 15, followed by a GDP (gemcitabine, cisplatin, and dexamethasone) regimen every three weeks. A rapid partial response (PR) to selinexor was observed within two weeks of treatment. Selinexor was found to be tolerable and safe. This patient reported mild hemocytopenia as the most common adverse reaction, without nausea, vomiting, or hyponatremia.[65]

Daratumumab

CD38 is a 48-kDa transmembrane glycoprotein that can be observed on the surface of many hematopoietic cells, such as multiple myeloma cells. It provides several functions, including receptor-mediated adhesion, signaling, and regulation of cyclase and hydrolase activity.[66,67]
Daratumumab is a human IgG1 monoclonal antibody that binds with high affinity to a unique CD38 epitope expressed on malignant cells and possesses direct and indirect antitumor activity and multiple mechanisms of action. Immune-mediated actions include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP); and immunomodulatory functions that target and deplete CD38-positive regulator immune suppressor cells, resulting in T-cell expansion and activation in patients with a response.[68,69]
Currently, daratumumab is under investigation for many types of hematological malignancies.
CD38 expression has been linked to a number of these diseases: in addition to multiple myeloma, consistent expression has been observed in the malignant cells of CLL, and it has also been reported in Waldenstrom macroglobulinemia, mantle cell lymphoma, acute lymphoblastic leukemia, acute myeloid leukemia, NK cell leukemia, and NK/T-cell lymphoma.[70-74]
Daratumumab induces the death of CD38-expressing tumor cells through multiple mechanisms, including complement-mediated cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) effects, antibody-dependent cellular phagocytosis (ADCP), apoptosis, and, to a lesser extent, inhibition of the enzymatic activity of CD38.[75-79]
Plasmablastic lymphomas are often CD38+ and share some biological and phenotypic features with multiple myeloma. Because of this, it seems reasonable to think that daratumumab could be used as proof of concept of activity in people with PBL who fail their first line of conventional chemotherapy or who are not eligible for autologous stem cell transplantation.
Table 4 summarizes studies of the use of daratumumab in PBL.


Table 4
Table 4. Literature review of Daratumumab therapy reports and Clinical Trial in PBL.

Ryu assessed the effectiveness and safety of daratumumab-based treatment plans in seven patients with advanced-stage PBL.[86] Out of these patients, 6 were considered evaluable, 4 had classic PBL, and 1 did not satisfy the precise criteria for PBL as established by the World Health Organization (WHO).[86] The initial assessment revealed that the median age of the patients included was 48 years. Additionally, all seven patients had disease locations outside the lymph nodes, 4 patients had ECOG scores of 3 or 4 at diagnosis, and 5 patients proved positive for CD38. Five patients underwent six cycles of daratumumab in conjunction with DA-EPOCH, and one patient underwent weekly daratumumab in addition to the addition of bortezomib. All patients who could be evaluated obtained a complete response (CR); four individuals were in remission after completing treatment. As of the data cutoff, the median duration of response (DOR) for the patients who could be evaluated was 16.8 months, and all of them were still showing a response. Patients diagnosed with classic PBL had a median duration of response (DOR) of 23.7 months, whereas patients who did not fulfill the stringent criteria for PBL had a median DOR of 3 months. The median overall survival (OS) and progression-free survival (PFS) were indeterminate in patients with classic PBL, except for one patient who died due to illicit drug use, asthma exacerbation, and respiratory arrest. In contrast, the median OS and PFS were 7 months and 6 months in patients who did not meet the strict criteria for PBL. Patients who failed to satisfy the stringent specifications for PBL experienced relapse and succumbed quickly after the completion of treatment. The 24-month overall survival rate for all patients who could be evaluated, regardless of whether they died from causes unrelated to treatment or disease, was 57%. Out of seven patients, six experienced severe adverse effects. There were no recorded fatalities associated with the treatment. Following the administration of daratumumab, two patients experienced infusion-related events, including a rash and itching in one patient and bradycardia in another.
In another small study, Daratumumab was also combined with the standard NHL salvage regimen, ICE. Five PBL patients were described.[41] Three patients were administered daratumumab in combination with EPOCH as their first-line treatment, while one patient received daratumumab together with lenalidomide, dexamethasone, and doxorubicin after experiencing a relapse. Three of the four patients maintained a state of remission for a minimum of 15 months after obtaining a complete response (CR).[41]
The AIDS Malignancy Consortium is now carrying out a limited, prospective investigation on the use of daratumumab-EPOCH in the first-line treatment of PBL (NCT04139304).
This study aims to assess the efficacy of daratumumab in combination with dose-adjusted etoposide, prednisone, vincristine sulfate, cyclophosphamide, and doxorubicin hydrochloride (DA-EPOCH) in the treatment of patients who have recently been diagnosed with stage I-IV plasmablastic lymphoma. The main goal is to determine the effectiveness of including daratumumab in the DA-EPOCH regimen by examining the proportion of patients with PBL who successfully complete at least three cycles of treatment according to the established protocol.
Daratumumab is administered to patients intravenously (IV) on days 1 (± 3 days), 8 (± 2 days), and 15 (± 2 days) during cycles 1-3, and on day 1 of cycles 4-6. Patients are given etoposide, doxorubicin hydrochloride, and vincristine sulfate intravenously for a continuous period of 96 hours on days 1-4. In addition, they orally provide prednisone for a period of 5 days, beginning on the first day, and intravenously administer cyclophosphamide for a length of 1 hour on the fifth day. The treatment is given at intervals of 21 days for a maximum of 6 cycles, unless there is a clear indication of disease recurrence or the patient experiences unacceptable adverse reactions.
Presently, another ongoing clinical study is assessing the efficacy of Daratumumab in conjunction with Bortezomib and dexamethasone for patients with refractory or recurrent PBL (NCT04915248).
This study is a phase II clinical trial conducted at many centers. It is an open-label, single-arm trial.
A total of 28 patients are expected to commence therapy over a period of 18 months, with recruitment taking place in 19 Italian FIL centers. The study's primary endpoint will be examined approximately 12 months after the final patient is enrolled, independent of the patient's response to the treatment outlined in the protocol. The study is expected to last around 2.5 years. Patients will be recruited based on their specific diagnosis and local assessment of CD38 expression at a level of 5%. The screening phase of the study involves conducting baseline assessments under local norms and study requirements. Induction treatment consists of one course (cycle 1) of daratumumab sc as a single agent, followed by eight courses (cycles 2–9) of daratumumab sc in combination with bortezomib sc and dexamethasone (DVd regimen). A maximum of 6 cycles (cycles 10-15) of daratumumab SC as a single agent will be administered to patients who achieve at least an SD after the induction phase. Every 21 days, induction cycles will be administered, while every 28 days, maintenance cycles will be administered.

Polatuzumab Vedotin

Nearly 40% of all cases of plasmablastic lymphoma have CD79a expression, ranging from 35% in HIV-negative patients to 45% in HIV-positive patients and 68% in post-transplant patients.[5-12] Thus, CD79a has been identified as a desirable therapeutic target for PBL.
Polatuzumab vedotin (Pola) is an innovative antibody-drug combination. It comprises a monoclonal antibody that targets CD79b, a component of the B-cell receptor found on normal B cells. This antibody is chemically linked to the microtubule-disrupting anti-mitotic agent, monomethyl auristatin.[89,90] A phase 1 trial demonstrated the safety of using polatuzumab vedotin alone in patients with severely treated B-cell malignancies, including non-Hodgkin lymphoma and chronic lymphocytic leukemia.[91,92] In real-world research, two patients with PBL were recently treated with a combination of polatuzumab, vedotin, and bendamustine. However, there is currently no available data on the outcomes of this treatment.[93] It is plausible that Pola could be used in a clinical trial for patients with relapsed or refractory PBL.

PD-1/PD-L1 Blocking Therapies

PD-1 binds to PD-L1 or PD-L2 on the surface of tumor cells and/or tumor-associated macrophages (TAM) in the tumor microenvironment, transmitting inhibitory signals to the T-cell receptor (TCR) pathway. Consequently, TCR-mediated signaling activation and cellular proliferation are inhibited.[94-101]
The inhibition of the PD-1/PD-L1 pathway can free T-cells from the inhibitory effects of tumor cells and restore the T-cell-mediated antitumor immune response. In recent years, significant progress has been made in developing cancer immunotherapies, such as PD1/PD-L1 inhibition. James Allison (MD Anderson Cancer Center, Houston, Texas, U.S.A.) and Tasuku Honjo (Kyoto University, Kyoto, Japan) were awarded the Nobel Prize in Physiology or Medicine in 2018 for their discoveries that made cancer immunotherapy possible.
PD-L1 levels expressed by tumor cells are generally associated with a response to PD-1/PD-L1 inhibitor therapies, widely used to treat patients with non-hematologic and hematologic malignancies, such as lung cancer, melanoma, and lymphoma. These inhibitors can prevent the binding of PD-1 to its ligands, thereby restoring the T-cell immune response and resulting in substantial and durable patient responses. The highest response rates have been observed in patients with classic Hodgkin lymphoma (CHL) among hematolymphoid neoplasms. In contrast, patients with non-Hodgkin lymphomas, including diffuse large B-cell lymphoma (DLBCL) and T-cell lymphomas with heterogeneous PD-L1/PD-L2 expression, have exhibited variable responses. Immunohistochemistry has been the primary method for assessing PD-L1 positivity in neoplastic cells based on PD-L1 expression. The positivity threshold values for PD-L1 vary between studies. For instance, a 5% cut-off was used in an early study, whereas different cut-offs were used in different lymphoma studies.[102-111]
An analysis of 82 patients with PBL revealed that almost all cases exhibited the presence of programmed death ligand 1 (PDL1) and programmed cell death protein 1 (PD1) in the immune infiltrate. Furthermore, a significant proportion of these cases, specifically one quarter, demonstrated substantial expression of PDL1 in both tumor cells and immune cells.[112]
In Epstein-Barr virus-positive PBL (EBV+ PBL), there was a higher level of overexpression of PD1/PDL1 in the microenvironment.[113]
Immunotherapy is thus becoming a viable treatment choice for this condition. A case study documented the use of nivolumab as salvage therapy in a patient with PDL1+ PBL, which successfully allowed an allogeneic stem cell transplant.[114]
A case study involving a patient with chemoresistant EBV + PBL showed that treatment with tislelizumab plus lenalidomide resulted in complete remission (CR) and an overall survival of 18 months.[115] Moreover, a recent case study documented a successful treatment outcome using pembrolizumab and radiotherapy in a patient with HIV-negative, EBV-positive recurrent plasmablastic lymphoma.[116]

Chimeric Antigen Receptor T-Cell Therapy

The field of therapeutic T cell engineering has gained significant attention recently because of the remarkable achievements of CD19 (chimeric antigen receptor) CAR treatment. Chimeric antigen receptors (CARs) are artificial receptors that alter the specificity and modify the function of T cells into which they are genetically inserted.[117-119]
The CD19 antigen is a member of the immunoglobulin superfamily and is specifically expressed in B-lymphocytes. Its expression is limited to the B-cell lineage, beginning in the early stages of B cell development, which coincide with heavy chain immunoglobulin rearrangement, and continuing until the later stages of B cell differentiation. Notably, the expression of CD19 increases as B cells differentiate. CD19, CD21, CD81, and Leuk-13.5 combine on the cell membrane of B-lymphoid cells to create a transduction complex. In addition, CD81 controls the level of CD19 expression during B cell growth.[117-119]
Over the past ten years, research has clearly shown and emphasized the important role of CD19-CAR-T cells in treating people with DLBCL that has relapsed or are not responding to treatment. The therapeutic function was designated for patients with a resistant disease and experience an early recurrence. CD19-CAR-T cells have demonstrated consistent therapeutic action in DLBCL patients with only a partial response after salvage therapy.[119-124]
CD19-CAR-T cells have effectively treated high-risk DLBCL patients as a first-line treatment. However, further research is necessary to evaluate their efficacy compared to traditional treatments. There isn't enough information yet to say which of the four commercially available CAR products for people with B cell lymphomas—Axi-Cel, Brexa-Cel, Liso-Cel, and Tisagenlecleucel—works best and causes the fewest side effects.[119-124]
Sadly, the majority of plasmablastic lymphomas lack CD19 expression. Nevertheless, only a small proportion of those patients can express this receptor, making CAR-T therapy plausible.
Raychaudhuri et al.[125] demonstrated that PBL can respond positively to axi-cel when CD19 is expressed. This patient's response was transient but clinically significant. Pain and transfusion-dependent cytopenias resolved, and the patient's performance status returned to normal. Longer in vivo persistence of axi-cell activity may have allowed for a more robust response.[125]

Autologous Bone Marrow Transplantation (ABMT)

Multiple studies have shown that up to 60 percent of patients with relapsed or refractory lymphoma will progress before ABMT.[126] In addition, the prognosis for patients with recurrent or resistant PBL is dismal.[127] Since the outcome appears significantly better in patients with relapsed NHL who received ABMT compared to those who did not, it seems reasonable to investigate the use of ABMT earlier in the course of disease, at least in the high-risk subgroup of patients.
A study using the EBMT registry found that some types of NHL, like plasmablastic lymphoma, had a higher risk of relapse compared to DLBCL (relative risk, 3.4%; 95% confidence interval, 1.1% to 10.4%; P.03) and a possible trend toward worse survival. It was hypothesized that ABMT in CR1 could be advantageous for this subgroup of patients.[128]
GICAT (Gruppo Italiano Cooperativo AIDS e Tumori) presented an interim analysis of a phase II multicenter trial of early consolidation with ABMT in HIV-positive patients with NHL and subsequently published their experience in HIV-positive patients with PBL.[129,130]
Four patients had PBL in this study. One patient was excluded from the study due to prolonged cytopenia during induction therapy (she is still in CR1 at +34 months). The last three patients got transplants with the BEAM (BCNU, etoposide, cytarabine, and melphalan) conditioning regimen in CR1 after induction therapy. At +24, +19, and +13 months, all of them are alive and in remission. The 2-year PFS and OS were 73% and 76%, respectively, across the entire series, with a median follow-up of 19.5 months (range: 4 to 65).[129,130]
In the Italian GICAT center, two additional HIV-positive patients with high-risk PBL were treated with ABMT as upfront consolidation following CHOP induction, and the results were reported separately. One patient is still alive in CR at +83 months, while the other died 4 months after transplantation due to progression.[130]
Similar to the Italian cooperative group and the Moffitt Cancer Center, the Center for International Blood and Marrow Transplant Research (CIBMTR) grouped patients with PBL by disease status. Seven of eleven (64%) patients who had ABMT in CR1 were still alive at the most recent follow-up, after an average of 25 months (range, 4 to 43 months); only four of nine (44%) patients who had ABMT in CR2 were still alive after an average of 62 months (range, 12 months). (CIBMTR, unpublished data). In a recent multicenter study involving 281 patients diagnosed with plasmablastic lymphoma, 13 participants had autologous bone marrow transplantation (ABMT) as a therapeutic consolidation following their initial complete remission. The current investigation lacks data regarding the responses or survival of these patients.[21] Ideally, randomized studies are required to establish the efficacy of ABMT in PBL. People with PBL who have any of the following high-risk factors should be considered for consolidation with ABMT as a first option: an aaIPI score greater than 2, no HIV, MYC gene rearrangement, TP53 gene deletion, or any response to induction chemotherapy other than CR (partial response or refractory disease).
 

Double Autologous Stem Cell Transplantation

There is only one report in the literature of an HIV-negative person with extraoral PBL who went into and stayed in complete remission for a very long time after intensive therapy with thalidomide and dexamethasone, followed by consolidation with two autologous stem cell transplants. The authors chose the intensive multiple myeloma-like treatment at random because antimyeloma drugs have been used in PBL case reports. The author stated that despite the inherent aggressiveness of the disease, the early presentation stage may have had a positive effect on the patient's long-term prognosis.[131]

Allotransplant for PBL

Compared to ABMT, the literature on allogeneic hematopoietic cell transplant (allo-HCT) PBL is considerably more limited. It's very important to know about the risks of opportunistic infections, having multiple infections at the same time (like viral hepatitis), the complicated drug interactions between antiretroviral drugs and transplant-related drugs, and how HIV affects T cell numbers and functions, the bone marrow microenvironment, and the cytokine milieu. These factors lead to both higher transplant-related mortality (TRM) and HIV-related mortality.
Because they lower TRM, reduced-intensity conditioning regimens have made allogeneic hematopoietic cell transplantation more successful overall, but it's still not clear what role they play in HIV-positive patients. A 51-year-old HIV-positive man with PBL and a hematopoietic cell transplant comorbidity index of 4 (high risk) underwent Allo HSCT from a matched unrelated donor to demonstrate the feasibility of the procedure.[132] He was given fludarabine, busulfan, and antithymocyte globulin to condition him. For the prevention of GVHD, methotrexate and tacrolimus were used. At the time of the report, he was two years post-transplantation, disease-free, and off immunosuppression.[132]
In a recent multicenter study involving 281 patients diagnosed with plasmablastic lymphoma, 5 participants had allo-HCT for relapsed PBL HIV negative. Out of these patients, four died away, with three deaths attributed to complications from the transplant and one death attributed to a second malignancy. One patient who received allo-HCT after one previous treatment is currently alive, as of the most recent follow-up, which occurred 26 months after the allo-HCT procedure. The patient is experiencing an important chronic graft-versus-host disease.[21]
Another report describing allo-HSCT in an HIV-negative PBL patient was found in the literature.[133] The patient experienced a recurrence following a consolidated ABMT. Then, he receives a salvage allo-HSCT from his daughter and achieves a favorable outcome. He has been in long-term, complete remission and is still alive. It may be attributable to his younger age, low IPI score, and prompt allo-HCT treatment following relapse. Thus, allo-HSCT has the potential to enhance the likelihood of long-term survival for young patients with PBL who have experienced a relapse.

Radiation Therapy (RT)

Radiotherapy is less considered a treatment option for PBL patients, as it has only been reported in approximately 200 published cases (Table 5).

Table 5
Table 5. Literature review of Radiation Therapy reports in PBL.

In 2024, systemic therapy will be used to treat most PBL cases. However, combined modality therapy (CMT), which includes systemic chemoimmunotherapy followed by consolidation radiation therapy (RT), is still a well-proven way to treat the disease. RT has multiple applications, either alone or in combination with multi-agent chemotherapy.[134-140]
After initial treatment, nearly half of PBL patients may experience either primary refractoriness or disease progression. Relapsed or refractory PBL remains exceedingly difficult to treat, with persistently dismal outcomes. Notably, RT still works well even for diseases that don't respond to chemotherapy. It has traditionally been an important part of clinical practice for these patients, with or without systemic therapy, as part of both curative and palliative-intent programs.[134-140]
Although radiation treatment (RT) remains a potential option for some patients who cannot undergo systemic therapy, it is primarily employed as a consolidation therapy following chemoimmunotherapy. Consolidation radiation is often recommended following multiple courses of systemic therapy in patients with advanced or bulky illness, several risk factors, or a partial response.[140-142]
Even in patients who achieve complete remission, the most common pattern of PBL relapse after chemotherapy involves the original sites of disease. RT may result in a benefit for event-free survival and, eventually, an overall survival (OS) benefit.[142-144]
Short courses of RT can alleviate a variety of symptoms, including pain, bleeding, airway or bowel obstruction, and neurologic compromise. Diseases that pose a threat to vital organs, such as the spinal cord or airway, may also be treated with RT to prevent impending complications. Finally, radiation therapy (RT) can be utilized as a potent treatment method for localized progression, with the aim of postponing the requirement for systemic therapy. This is particularly beneficial, as systemic therapy is often linked to a greater degree of adverse effects. The ideal doses for palliating R/R DLBCL are still unknown, and the most suitable treatment regimen may eventually vary depending on the specific clinical situation. Typically, hypofractionated doses ranging from 20 to 30 Gy are delivered. Various strategies may be suitable, depending on the clinical situation. Patients with a limited life expectancy are recommended to undergo short treatment regimens, such as receiving 4 Gy of radiation over 5 days, 8 Gy in a single day, or even 2 Gy over 2 days or 4 Gy in one day. Extended treatment schedules (e.g., 3 Gy for 10 days or 2.5 Gy for 15 days) may be more suitable for patients with a more positive outlook, particularly those with a smaller amount of disease. Generally, it is advisable to restrict therapy volumes to the bulk of the disease with the smallest possible margin.[142-148]

CNS Prophylaxis

CNS relapse is a relatively uncommon but frequently devastating complication of DLBCL. Most CNS relapses occur during or shortly after first-line immune-chemotherapy, with a median time of 6 to 8 months, as reported in a recent prospective clinical trial.[149]
Treating secondary CNS lymphoma (SCNSL) is sometimes challenging, and historically, the outcomes have been unsatisfactory. Consequently, significant attention has been given to identifying patients with the highest propensity to this problem, as well as implementing preventive measures aimed at minimizing risk as much as feasible. A better understanding of DLBCL's molecular biology and the CNS-IPI238 trial have helped find people at high risk for secondary central nervous system lymphoma (SCNSL). However, decisions about preventing this disease are still based on looking at past cases or extrapolating data from other types of the disease.[150-152] There have been no prospective randomized trials conducted to directly assess the effectiveness of CNS prophylaxis. Physicians often encounter the difficult task of preventing a potential consequence without exposing the patient to further treatment that may have harmful side effects and lack substantial evidence of its effectiveness.
Numerous studies have investigated possible CNS relapse risk factors in DLBCL. In 2016, the German High-Grade Non-Hodgkin Lymphoma Study Group (DSHNHL) created a prognostic model (CNS-IPI) that sorts patients into three risk groups based on the five standard IPI factors as well as whether the disease has spread to the kidneys or adrenal glands.[150,152]
Significantly, patients who had five or six risk variables had a respective probability of CNS relapse of 15% and 32.5%. Specific extranodal (EN) locations have been linked to a higher likelihood of central nervous system (CNS) recurrence. The CNS-IPI model considers the involvement of the kidneys and adrenal glands. However, intravascular lymphoma is known to have a high risk of involving the CNS, either at the start or during a relapse. The association between testicular involvement and CNS recurrence probability, ranging from 10% to 25% over a 10-year period, has been well-established in both limited and advanced stages.[150-154]
In a retrospective series, breast involvement was associated with a higher risk of CNS relapse (15%), whereas other EN sites, such as the uterus, blood, bone marrow, and epidural area, exhibited inconsistent results and are unlikely to be independently predictive of CNS relapse.[155]
A new systematic review looked at stand-alone IT prophylaxis in 7357 patients who were getting chemoimmunotherapy. The review included three post-hoc trial analyses and 10 retrospective investigations.[156] In univariable or multivariable analyses, IT prophylaxis was not associated with a reduction in CNS relapse rate. The administration of IT therapy can be difficult and uncomfortable for the patient, with some evidence suggesting an association with hospitalization for infection-related reasons in older patients.[150-157]
The utilization of brain imaging and lumbar puncture/cerebrospinal fluid (CSF) analysis to detect individuals at high risk of central nervous system (CNS) involvement who could potentially benefit from treatments targeting the CNS is increasing.[158]
Several studies have demonstrated that flow cytometric analysis of CSF is more sensitive than cytology for detecting occult CNS involvement.[159,161] Nonetheless, a percentage of patients with negative flow cytometry result in CNS relapse shortly after treatment, indicating the need for more sensitive techniques. The incidence of CNS relapse is estimated to occur in approximately 3–5% of PBL patients.[3-10]
Further, PBL in people living with HIV (PLWH) has an increased risk of aggressive disease, with CNS involvement occurring more frequently than other extra-nodal involvement.
886 people who were newly diagnosed with AIDS-related lymphomas (DLBCL and BL) were looked at in detail.[162] It was found that the central nervous system (CNS) was involved in between 5% and 30% of the cases at the start of the therapy. The recurrence of central nervous system (CNS) cancer happens promptly, typically within a median period of 4.2 months following diagnosis, and is associated with a very poor survival rate of only 1.6 months. Over 90% of patients underwent intrathecal (IT) and central nervous system (CNS) prophylaxis, while 5% encountered CNS recurrence. In this case, it has not been determined what the best treatment plan, dosage frequency, and ways to avoid complications in the central nervous system (CNS), such as choosing between intravenous medications that can reach the CNS and intrathecal therapy. The guidelines from the US National Comprehensive Cancer Network recomment the use of intrathecal methotrexate for central nervous system prophylaxis in all people living with HIV (PLWH) who have lymphoma.[22]
In conclusion, people with PBL are likely to get leptomeningeal disease because of the high rate of proliferation, the strong link to HIV infection, the high rate of extranodal involvement, and the presence of MYC translocations. CNS prophylaxis should be considered in a case-by-case basis.

Antiretroviral Treatment During Chemotherapy for HIV+ PBL

It is crucial to note that more than 60% of individuals diagnosed with plasmablastic lymphoma are also infected with HIV. Combined antiretroviral therapy (cART) has independently contributed to improving the response to chemotherapy and the survival of HIV-infected patients with lymphoma.[163-166]
All HIV-positive patients with PBL should, therefore, receive cART concurrently with chemotherapy. The antiretroviral treatment history, HIV strain sensitivity, HLAB5701 result, and hepatitis B virus (HBV) infection markers should be considered when choosing a cART regimen. In certain instances, it is recommended to obtain the HIV strain's tropism result (R5, X4, or dual tropism). In addition, it is essential to always consider the potential pharmacological interactions and cross-toxicity between antiretrovirals and antitumor drugs or other commonly used drugs in this patient population, such as antifungals. There is limited clinical evidence regarding the efficacy and safety of chemotherapy and antiretroviral therapy.
In a series of 150 HIV-infected patients with cancer (mostly hematological malignancies), protease inhibitor-based cART regimens were less effective and less safe than those based on non-nucleoside reverse transcriptase inhibitors (NNRTI) and integrase strand transfer inhibitors (ISTI) (INSTI).[167] Between 40 and 60 percent of HIV-infected patients have been exposed to HBV, and between 3 and 10 percent have chronic hepatitis B, which is defined by the presence of HBV surface antigen (HBsAg).[168-172] Reactivation of HBV can occur during chemotherapy, particularly when rituximab is employed as a therapeutic agent.[172-175] Patients co-infected with HIV and HBV should receive a combination antiretroviral therapy (cART) regimen that is also effective against HBV. In addition to tenofovir and emtricitabine (FTC) or lamivudine (3TC), a third HIV drug should be administered.[174,175] Tenofovir alafenamide (TAF) should be preferred over tenofovir disoproxilfumarate (TDF) because it has a more favorable safety profile and is equally effective against HBV.[175] Therefore, these antiretroviral drugs can alter the pharmacokinetics of antitumor drugs that are substrates of this isoenzyme, particularly taxanes and alkylating agents like cyclophosphamide and etoposide and, to a lesser extent, vinca alkaloids, antitumor antibiotics, and platinum. There are no significant interactions between antiretrovirals and anthracyclines because aldose reductase metabolizes both substances. In patients receiving concomitant CHOP, doxorubicin, etoposide, and PI-based cART, cyclophosphamide clearance was decreased, and the frequency of severe anemia and neutropenia was increased compared to patients receiving CHOP alone. Severe cases of neutropenia and mucositis were reported in patients receiving concomitant CHOP and cART.[176]
It is unlikely that nucleoside or nucleotide reverse transcriptase inhibitors (NRTIs) will interact pharmacokinetically with cytostatics. The CCR5 antagonist maraviroc is a substrate for the CYP3A enzyme and the P glycoprotein. This means that strong CYP3A inducers or inhibitors may change the levels of maraviroc. Insti (raltegravir and dolutegravir) metabolizes through glucuronidation in the liver and has minimal interactions with cytostatic medications. Elvitegravir, a third member of this class, must be co-administered with the cobicistat enhancer so that their interactions are functionally equivalent to those of the PI. No pharmacokinetic interactions between antiretrovirals and rituximab, the most widely used monoclonal antibody for NHL, have been described.[175-180]
When selecting a combination antiretroviral therapy (cART) regimen, it is also crucial to consider the drugs' safety profile. 3TC, FTC, abacavir (AUC), and TDF or TAF are the most commonly used NRTIs for the treatment of HIV at present. 3TC, FTC, TDF, and TAF are also effective anti-HBV agents. 3TC, FTC, and ABC are not cross-toxic with anticancer drugs. TDF can cause proximal tubular nephropathy and a decreased glomerular filtration rate; therefore, patients with renal insufficiency, tumor lysis syndrome, or those receiving antitumor drugs with nephrotoxic potential should avoid using TDF. TAF is equivalent to TDF in terms of efficacy and lacks nephrotoxicity, at least in the short term.[181,182] Other NRTIs, such as zidovudine (AZT), didanosine (ddI), and stavudine (d4T), are rarely used today and have significant cross-toxicity with certain anti-tumor drugs. AZT has the potential to be myelotoxic and may increase the hematological toxicity of various chemotherapeutic regimens. Certain antitumor medications, such as platinum, taxanes, and vinca alkaloids, can worsen peripheral neuropathy caused by DdI and d4T and cause mitochondrial toxicity.[183] Some PIs, like atazanavir, lopinavir, and saquinavir, may make the CT interval longer. This is something that should be thought about when these drugs are combined with anthracyclines, which are also known to make the CT interval longer. The bilirubin levels of patients with hepatopathy can be used to adjust the dosage of certain antitumor drugs. Although atazanavir can cause unconjugated hyperbilirubinemia because it blocks the uridine diphosphate glucuronosyltransferase 1A1 enzyme (UGT1A1), this can make it harder to make the necessary changes when taking this antiretroviral.[184]

Complementary Treatments

At the time of diagnosis or after beginning chemotherapy, patients with a large tumor mass may exhibit complications resulting from tumor lysis syndrome. In cases where this complication is likely because of the size of the tumor or very high levels of LDH and uric acid, hyperhydration, forced diuresis, and allopurinol should be given before chemotherapy. Additionally available is rasburicase, a recombinant version of the urate oxidase enzyme that turns uric acid into allantoin, which the kidneys excrete more effectively. This medication is more effective than allopurinol at reducing plasma uric acid levels and can prevent chemotherapy from beginning too late.[185,186] The recommended intravenous dose of rasburicase is 0.20 mg/kg/day in 50 mL of normal saline for 30 minutes. The duration of treatment ranges between 5 and 7 days, but shorter-duration regimens appear to be equally effective. Giving granulocyte colony-stimulating factors (G-CSF) is suggested to shorten the time of neutropenia after chemotherapy, improve cytostatic tolerance, and allow full doses and proper chemotherapy intervals.[187]

Prophylaxis of Opportunistic Infections Associated with HIV+PBL

After the administration of chemotherapy, the total number of CD4+ T lymphocytes decreases by 30–50% with respect to the baseline, depending on the intensity of the treatment and the moment at which the analysis is carried out. This is why the risk of opportunistic infections associated with HIV is higher in these patients than in patients with PBL at a similar stage.[188]
In principle, it should be said that primary or secondary prophylaxis indicated according to the CD4+ T lymphocyte count and the previous history of opportunistic infections should be performed. However, it is recommended to consider that the degree of immunosuppression in patients is greater than the one shown by the CD4+ T lymphocyte count at the time of tumor diagnosis and that these lymphocyte markers should be monitored throughout the treatment of the lymphoma and act accordingly.[189]
Pneumocystis jirovecii prophylaxis is recommended for all patients. However, we must consider the effects that systematic implementation of this practice can have on antibiotic resistance and other undesirable outcomes, such as Clostridium difficile colitis.[189-190] Anti-CMV prophylaxis is generally not recommended, but close monitoring with periodic blood PCR determinations for this virus is recommended in those patients with a low CD4+ T lymphocyte count (every 7 days).

Vaccinations

The annual inactivated influenza vaccination and COVID-19 are recommended for both HIV-positive and HIV-negative PBL patients.
The COVID-19 vaccine is particularly important for those patients due to the frequent prolonged positivity and virus shedding of the SARS-Cov-2 and the impact of this on delaying the chemotherapy.[191,192] The vaccination of close contacts against influenza and COVID-19 is also recommended. 278 As with all HIV-infected patients, PBL patients should also receive vaccinations against pneumococcus, HBV, and the hepatitis A virus. Although the optimal time to administer the vaccines is unknown, it is recommended to do so at least two weeks prior to beginning chemotherapy or at least one week after the last cycle.[193,194]

First Line Post-Treatment Assessment

After the initial treatment, if patients achieve a PET-negative remission, we schedule additional monitoring. Regrettably, even after discontinuation of cytotoxic treatment and achieving complete remission, numerous patients have fatigue, polyneuropathy, or anxiety. Thus, we promptly direct patients experiencing anxiety to receive psychological treatment and, if judged required, commence the administration of psychotropic medications. For patients diagnosed with polyneuropathy, we advise decreasing the dosage of vincristine and providing symptomatic relief using gabapentin or duloxetine, even though there is limited evidence supporting their effectiveness. Moreover, cancer rehabilitation clinics recommend that patients reside there for a period of time to improve their overall quality of life. For additional monitoring, we refrain from using routine CT scans on asymptomatic individuals because the American Society of Hematology has determined that they are not effective. We suggest performing a blood count, renal and liver function tests, LDH measurement, and a clinical examination every 3 months during the initial 2-year period following treatment. Patients with prominent mediastinal or retroperitoneal disease at diagnosis should undergo chest X-rays and ultrasonography. The NCCN guidelines continue to recommend surveillance CT scans for follow-up in patients with aggressive lymphoma, although we only utilize this approach for specific patients who have an increased risk of experiencing a relapse.

Relapsed/Refractory Plasmablastic Lymphoma

In patients with persistent PET-positive disease, new lymphadenopathy, or organ lesions after first-line treatment, we perform a new biopsy to confirm malignancy and the former diagnosis. Refractory PBL has a significantly worse prognosis than other aggressive non-Hodgkin B-cell lymphomas, irrespective of whether the patient has HIV or not. The reported median PFS ranges from 6–7 months, while the median OS ranges from 11–13 months.[5-12]
However, due to the rarity of this disease, there is insufficient evidence to support one particular salvage therapy for patients with relapsed or refractory disease, particularly for those who do not achieve at least a partial response. Furthermore, the same treatment methods used as the first line of therapy can also be employed in later stages of treatment. If accessible, we suggest enrolling these patients in experimental clinical trials. At the moment, there aren't many prospective randomized studies that directly compare the different second-line treatment options for people with PBL who can't get autologous stem cell transplantation (ASCT) or anti-CD19 CAR T-cell therapy. Such a comparison would yield valuable data for treatment sequencing and evaluate treatment effectiveness in various patient subgroups, including those with high clinical and biological risk factors. Managing these patients in this context is currently a significant unmet medical need. However, situations where randomized studies are scarce are increasingly seeing prevalent actual comparisons across different groups, particularly those that are carefully matched.
Ultimately, we have created a comprehensive table that aims to condense the complex therapy recommendations for this uncommon and aggressive lymphoma (Figure 1).

Figure 1
Figure 1. Complex therapy recommendations for Relapsed/Refractory Plasmablastic Lymphoma.


Conclusions

In terms of both diagnosis and treatment, PBL is a challenging disease. The majority of long-term survivors had limited disease or were eligible for autologous stem cell transplants as consolidation following combination chemotherapy responses. Regimens for myeloma that include proteasome inhibitors, immunomodulators, and targeted therapy pave the way for improved outcomes. Now that we have gained more knowledge about the mutational landscape of PBL, researchers have suggested numerous potential new targets. These include pan-TRK inhibitors like larotrectinib or entrectinib for NTRK3 mutations. It's also important to look at other effective treatments that have been used for MM, like CAR-T and bispecific antibodies against CD38, CD138, or B-cell maturation antigen (BCMA), especially for patients with relapsed and resistant PBL.  

References   

  1. Stein H. & Dallenbach F. in Neoplastic Hematopathology (ed.Knowles, D.M.) 675-714 (Williams & Wilkins, Baltimore, MA, 1992).
  2. Delecluse HJ, Anagnostopoulos I, Dallenbach F, et al. Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection. Blood. 1997;89:1413-1420. https://doi.org/10.1182/blood.V89.4.1413 PMid:9028965
  3. Bibas M. Plasmablastic Lymphoma. A State-of-the-Art Review: Part 1-Epidemiology, Pathogenesis, Clinicopathologic Characteristics, Differential Diagnosis, Prognostic Factors, and Special Populations. Mediterr J Hematol Infect Dis. 2024 Jan 1;16(1):e2024007. https://doi.org/10.4084/MJHID.2024.007 PMid:38223486 PMCid:PMC10786126
  4. 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 Jul;36(7):1720-1748. https://doi.org/10.1038/s41375-022-01620-2 PMid:35732829 PMCid:PMC9214472
  5. 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 Sep 15;140(11):1229-1253. https://doi.org/10.1182/blood.2022015851 PMid:35653592 PMCid:PMC9479027
  6. Castillo JJ, Bibas M, Miranda RN. The biology and treatment of plasmablastic lymphoma. Blood. 2015 Apr 9;125(15):2323-30. https://doi.org/10.1182/blood-2014-10-567479 PMid:25636338
  7. Bibas M, Castillo JJ. Current knowledge on HIVassociated Plasmablastic Lymphoma. Mediterr J Hematol Infect Dis. 2014;6(1):e2014064. https://doi.org/10.4084/mjhid.2014.064 PMid:25408850 PMCid:PMC4235470
  8. Castillo JJ, Furman M, Beltrán BE,et al.: Human immunodeficiency virus-associated plasmablastic lymphoma: poor prognosis in the era of highly active antiretroviral therapy. Cancer. 2012 Nov 1;118(21):5270-7. https://doi.org/10.1002/cncr.27551 PMid:22510767
  9. Florindez JA, Alderuccio JP, Reis IM, Lossos IS. Survival analysis in treated plasmablastic lymphoma patients: a population-based study. Am J Hematol. Nov 2020;95(11):1344-1351. https://doi.org/10.1002/ajh.25955 PMid:32777103
  10. Alec R. Hansen , Victoria A. Vardell , Lindsey A. Fitzgerald , Epidemiologic characteristics, treatment patterns, and survival analysis of Plasmablastic Lymphoma in the US: A SEER and NCDB analysis., Clinical Lymphoma, Myeloma and Leukemia (2023), https://doi.org/10.1016/j.clml.2023.12.014 PMid:38262787
  11. Castillo J, Pantanowitz L, Dezube BJ. HIV-associated plasmablastic lymphoma: lessons learned from 112 published cases. Am J Hematol. 2008 Oct;83(10):804-9. https://doi.org/10.1002/ajh.21250 PMid:18756521
  12. Castillo JJ, Winer ES, Stachurski D, Perez K, Jabbour M, Milani C, Colvin G, Butera JN. Prognostic factors in chemotherapy-treated patients with HIV-associated Plasmablastic lymphoma. Oncologist. 2010;15(3):293-9. https://doi.org/10.1634/theoncologist.2009-0304 PMid:20167839 PMCid:PMC3227958
  13. Ibrahim IF, Shapiro GA, Naina HVK. Treatment of HIV-associated plasmablastic lymphoma: a single-center experience with 25 patients. J Clin Oncol 2014; 32(15Suppl):8583. https://doi.org/10.1200/jco.2014.32.15_suppl.8583
  14. Loghavi S, Alayed K, Aladily TN, Zuo Z, Ng SB, Tang G, Hu S, Yin CC, Miranda RN, Medeiros LJ, Khoury JD. Stage, age, and EBV status impact outcomes of plasmablastic lymphoma patients: a clinicopathologic analysis of 61 patients. J Hematol Oncol. 2015 Jun 10;8:65. https://doi.org/10.1186/s13045-015-0163-z PMid:26055271 PMCid:PMC4472407
  15. Pinnix CC, Shah JJ, Chuang H, et al. Doxorubicin-based chemotherapy and radiation therapy produces favorable outcomes in limited-stage plasmablastic lymphoma: a single-institution review. Clin Lymphoma Myeloma Leuk 2016; 16:122-8. https://doi.org/10.1016/j.clml.2015.12.008 PMid:26795083 PMCid:PMC9754636
  16. Rudresha AH, Lakshmaiah KC, Agarwal A, et al. Plasmablastic lymphoma in immunocompetent and in immunocompromised patients: experience at a regional cancer centre in India. South Asian J Cancer 2017; 6:69-71. https://doi.org/10.4103/sajc.sajc_186_16 PMid:28702410 PMCid:PMC5506813
  17. Focà E, Cavaglià G, Rusconi S, et al. Survival in HIV-infected patients with lymphoma according to the choice of antiretroviral treatment: an observational multicentre study. HIV Med 2018, Online ahead of print https://doi.org/10.1111/hiv.12624 PMid:29862615
  18. Al Tabaa Y, Tchernonog E, Faurie P, et al. Post-treatment positron emission tomography-computed tomography is highly predictive of outcome in plasmablastic lymphoma. Eur J Nucl Med Mol Imaging 2018; 45:1705-9. https://doi.org/10.1007/s00259-018-4020-5 PMid:29679112
  19. Zuze T, Painschab MS, Seguin R, et al. Plasmablastic lymphoma in Malawi. Infect Agent Cancer 2018; 13:22. https://doi.org/10.1186/s13027-018-0195-4 PMid:29988350 PMCid:PMC6022505
  20. Rodrigues-Fernandes CI, de Souza LL, Dos Santos-Costa SF, et al. Clinicopathological analysis of oral plasmablastic lymphoma: a systematic review. J Oral Pathol Med 2018; 47:915-22. https://doi.org/10.1111/jop.12753 PMid:29917262
  21. Di Ciaccio PR, Polizzotto MN, Cwynarski K, Gerrie AS, Burton C, Bower M, Kuruvilla J, Montoto S, McKay P, Fox CP, Milliken S, Jiamsakul A, Osborne W, Collins GP, Manos K, Linton KM, Iyengar S, Kassam S, Limei MP, Kliman D, Wong Doo N, Watson AM, Fedele P, Yannakou CK, Hunt S, Ku M, Sehn LH, Smith A, Renshaw H, Maxwell A, Liu Q, Dhairyawan R, Ferguson G, Pickard K, Painter D, Thakrar N, Song KW, Hamad N. The influence of immunodeficiency, disease features, and patient characteristics on survival in plasmablastic lymphoma. Blood. 2024 Jan 11;143(2):152-165. https://doi.org/10.1182/blood.2023021348 PMid:37832030
  22. NCCN Clinical Practice Guidelines in Oncology: B-Cell Lymphomas. NCCN 2024;Version 1..2024- 18 January 2024
  23. Robak P, Robak T. Bortezomib for the Treatment of Hematologic Malignancies: 15 Years Later. Drugs R D. 2019 Jun;19(2):73-92. https://doi.org/10.1007/s40268-019-0269-9 PMid:30993606 PMCid:PMC6544598
  24. Leonard JP, Kolibaba K, Reeves JA, et al. Randomized phase 2 openlabel study of R-CHOP ± bortezomib in patients (Pts) with untreated non-germinal center B-cell-like (Non-GCB) subtype diffuse large cell lymphoma (DLBCL): results from the Pyramid trial (NCT00931918). Blood. 2015;126(23):811. 44. https://doi.org/10.1182/blood.V126.23.811.811
  25. Robak T, Huang H, Jin J, et al. Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N Engl J Med. 2015;372(10):944-953. https://doi.org/10.1056/NEJMoa1412096 PMid:25738670
  26. Dunleavy K, Pittaluga S, Czuczman MS, Dave SS, Wright G, Grant N, Shovlin M, Jaffe ES, Janik JE, Staudt LM, Wilson WH. Differential efficacy of bortezomib plus chemotherapy within molecular subtypes of diffuse large B-cell lymphoma. Blood. 2009 Jun 11;113(24):6069-76. https://doi.org/10.1182/blood-2009-01-199679 PMid:19380866 PMCid:PMC2699229
  27. Jambusaria A, Shafer D, Wu H, Al-Saleem T, Perlis C. Cutaneous plasmablastic lymphoma. J Am Acad Dermatol. 2008 Apr;58(4):676-8. https://doi.org/10.1016/j.jaad.2007.08.009 PMid:18342714
  28. Bose P, Thompson C, Gandhi D, Ghabach B, Ozer H. AIDS-related plasmablastic lymphoma with dramatic, early response to bortezomib. Eur J Haematol. 2009 Jun;82(6):490-2. https://doi.org/10.1111/j.1600-0609.2009.01235.x PMid:19220417
  29. Bibas M, Grisetti S, Alba L, Picchi G, Del Nonno F, Antinori A. Patient with HIV-associated plasmablastic lymphoma responding to bortezomib alone and in combination with dexamethasone, gemcitabine, oxaliplatin, cytarabine, and pegfilgrastim chemotherapy and lenalidomide alone. J Clin Oncol. 2010 Dec 1;28(34):e704-8. https://doi.org/10.1200/JCO.2010.30.0038 PMid:20823416
  30. Lipstein M, O'Connor O, Montanari F, Paoluzzi L, Bongero D, Bhagat G. Bortezomib-induced tumor lysis syndrome in a patient with HIV-negative plasmablastic lymphoma. Clin Lymphoma Myeloma Leuk. 2010 Oct;10(5):E43-6. https://doi.org/10.3816/CLML.2010.n.074 PMid:21856550
  31. Dasanu CA, Bauer F, Codreanu I, Padmanabhan P, Rampurwala M. Plasmablastic haemato-lymphoid neoplasm with a complex genetic signature of Burkitt lymphoma responding to bortezomib. Hematol Oncol. 2013 Sep;31(3):164-6. https://doi.org/10.1002/hon.2024 PMid:22899491
  32. Saba NS, Dang D, Saba J, Cao C, Janbain M, Maalouf B, Safah H. Bortezomib in plasmablastic lymphoma: a case report and review of the literature. Onkologie. 2013;36(5):287-91. https://doi.org/10.1159/000350325 PMid:23689224
  33. Cao C, Liu T, Zhu H, Wang L, Kai S, Xiang B. Bortezomib-contained chemotherapy and thalidomide combined with CHOP (Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone) play promising roles in plasmablastic lymphoma: a case report and literature review. Clin Lymphoma Myeloma Leuk. 2014 Oct;14(5):e145-50. https://doi.org/10.1016/j.clml.2014.03.002 PMid:25225082
  34. Cencini E, Fabbri A, Guerrini S, Mazzei MA, Rossi V, Bocchia M. Long-term remission in a case of plasmablastic lymphoma treated with COMP (cyclophosphamide, liposomal doxorubicin, vincristine, prednisone) and bortezomib. Eur J Haematol. 2016 Jun;96(6):650-654. https://doi.org/10.1111/ejh.12732 PMid:26715026
  35. Hirosawa M, Morimoto H, Shibuya R, Shimajiri S, Tsukada J. A striking response of plasmablastic lymphoma of the oral cavity to bortezomib: a case report. Biomark Res. 2015 Nov 4;3:28. https://doi.org/10.1186/s40364-015-0053-0 PMid:26543559 PMCid:PMC4634743
  36. Castillo JJ, Reagan JL, Sikov WM, Winer ES. Bortezomib in combination with infusional dose-adjusted EPOCH for the treatment of plasmablastic lymphoma. Br J Haematol. 2015;169(3):352-355 https://doi.org/10.1111/bjh.13300 PMid:25612847
  37. Fernandez-Alvarez R, Gonzalez-Rodriguez AP, Rubio-Castro A, et al. Bortezomib plus CHOP for the treatment of HIV-associated plasmablastic lymphoma: clinical experience in three patients. Leuk Lymphoma. 2016;57(2):463-466. https://doi.org/10.3109/10428194.2015.1050666 PMid:25976108
  38. Fedele PL, Gregory GP, Gilbertson M, Shortt J, Kumar B, Opat S, Grigoriadis G. Infusional dose-adjusted epoch plus bortezomib for the treatment of plasmablastic lymphoma. Ann Hematol. 2016 Mar;95(4):667-8. https://doi.org/10.1007/s00277-016-2601-6 PMid:26801792
  39. Arora N, Gupta A, Sadeghi N. Durable complete remission with combination chemotherapy and bortezomib in HIV-associated plasmablastic lymphoma. BMJ Case Rep. 2017 Oct 9;2017:bcr2017222063. https://doi.org/10.1136/bcr-2017-222063 PMid:28993364 PMCid:PMC5652596
  40. Guerrero-Garcia TA, Mogollon RJ, Castillo JJ. Bortezomib in plasmablastic lymphoma: a glimpse of hope for a hard-to-treat disease. Leuk Res. 2017;62(September):12-16 https://doi.org/10.1016/j.leukres.2017.09.020 PMid:28963907
  41. Dittus C, Grover N, Ellsworth S, Tan X, Park SI. Bortezomib in combination with dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin) induces long-term survival in patients with plasmablastic lymphoma: a retrospective analysis. Leuk Lymphoma. 2018 Sep;59(9):2121-2127. https://doi.org/10.1080/10428194.2017.1416365 PMid:29303024
  42. Castillo JJ, Guerrero-Garcia T, Baldini F, Tchernonog E, Cartron G, Ninkovic S, Cwynarski K, Dierickx D, Tousseyn T, Lansigan F, Linnik Y, Mogollon R, Navarro JT, Olszewski AJ, Reagan JL, Fedele P, Gilbertson M, Grigoriadis G, Bibas M. Bortezomib plus EPOCH is effective as frontline treatment in patients with plasmablastic lymphoma. Br J Haematol. 2019 Feb;184(4):679-682. https://doi.org/10.1111/bjh.15156 PMid:29527667
  43. Ando K, Imaizumi Y, Kobayashi Y, Niino D, Hourai M, Sato S, Sawayama Y, Hata T, Ohshima K, Miyazaki Y. Bortezomib- and Lenalidomide-Based Treatment of Refractory Plasmablastic Lymphoma. Oncol Res Treat. 2020;43(3):112-116. https://doi.org/10.1159/000504608 PMid:31842017
  44. Umeanaeto O, Gamboa J, Diaz J, Hakim MN, Corral J, Philipovskiy A, Gaur S. Incorporating Bortezomib in the Management of Plasmablastic Lymphoma. Anticancer Res. 2019 Sep;39(9):5003-5007. https://doi.org/10.21873/anticanres.13690 PMid:31519607
  45. Cai J, Qiu L, Ma L, Zhang N, Fan FY. Case Report: Bortezomib Plus CDOP Followed by Sequential Autologous Hematopoietic Stem Cell Transplantation and Lenalidomide-Based Maintenance Therapy in Plasmablastic Lymphoma. Front Med (Lausanne). 2021 Dec 3;8:749863. https://doi.org/10.3389/fmed.2021.749863 PMid:34926499 PMCid:PMC8677941
  46. Sabry W, Wu Y, Kodad SG. Bortezomib, Lenalidomide and Dexamethasone Combination Induced Complete Remission in Relapsed/Refractory Plasmablastic Lymphoma: Case Report of a Potential Novel Treatment Approach. Curr Oncol. 2022 Jul 18;29(7):5042-5053. https://doi.org/10.3390/curroncol29070399 PMid:35877259 PMCid:PMC9323819
  47. Gribben JG, Fowler N, Morschhauser F. Mechanisms of Action of Lenalidomide in B-Cell Non-Hodgkin Lymphoma. J Clin Oncol. 2015 Sep 1;33(25):2803-11. https://doi.org/10.1200/JCO.2014.59.5363 PMid:26195701 PMCid:PMC5320950
  48. Carras, S.; Regny, C.; Peoc'h, M.; Gervasoni, J.; Gressin, R.; Cahn, J.Y.; Molina, L. Dramatic efficacy of low dose lenalidomide as single agent in a patient with refractory gastric nonhuman immunodeficiency virus-associated plasmablastic lymphoma. Leuk. Lymphoma 2015, 56, 2986-2988. [CrossRef] [PubMed] https://doi.org/10.3109/10428194.2015.1016931 PMid:25676034
  49. Yanamandra, U.; Sahu, K.K.; Jain, N.; Prakash, G.; Saikia, U.; Malhotra, P. Plasmablastic lymphoma: Successful management with CHOP and lenalidomide in resource constraint settings. Ann. Hematol. 2016, 95, 1715-1717. https://doi.org/10.1007/s00277-016-2732-9 PMid:27324386
  50. Schmit J.M.; Delaune, J.; Norkin, M.; Grosbach, A. A case of plasmablastic lymphoma achieving complete response and durable remission after lenalidomide-based therapy. Oncol. Res. Treat. 2017, 40, 46-48. https://doi.org/10.1159/000455146 PMid:28095384
  51. Marrero, W.D.; Cruz-Chacón, A.; Castillo, C.; Cabanillas, F. Successful Use of Bortezomib-Lenalidomide Combination as Treatment for a Patient with Plasmablastic Lymphoma. Clin. Lymphoma Myeloma Leuk. 2018, 18, e275-e277. https://doi.org/10.1016/j.clml.2018.04.011 PMid:29753690
  52. Cheng, L.; Song, Q.; Liu, M.; Wang, Y.; Yi, H.; Qian, Y.; Xu, P.; Cheng, S.; Wang, C.; Wang, L.; et al. Case Report: Successful Management of a Refractory Plasmablastic Lymphoma Patient with Tislelizumab and Lenalidomide. Front. Immunol. 2021, 12, 702593. https://doi.org/10.3389/fimmu.2021.702593 PMid:34322131 PMCid:PMC8312258
  53. Lee M, Martin BA, Abdulhaq H. Daratumumab, Lenalidomide, and Dexamethasone (DRD), an Active Regimen in the Treatment of Immunosuppression-Associated Plasmablastic Lymphoma (PBL) in the Setting of Gorham's Lymphangiomatosis: Review of the Literature. Case Rep Hematol. 2022 Jun 27;2022:8331766. https://doi.org/10.1155/2022/8331766  PMid:35795542 PMCid:PMC9252825
  54. Yi JH, Kim SJ, Kim WS. Brentuximab vedotin: clinical updates and practical guidance. Blood Res. 2017 Dec;52(4):243-253. https://doi.org/10.5045/br.2017.52.4.243 PMid:29333400 PMCid:PMC5762734
  55. Donato EM, Fernández-Zarzoso M, Hueso JA, de la Rubia J. Brentuximab vedotin in Hodgkin lymphoma and anaplastic large-cell lymphoma: an evidence-based review. Onco Targets Ther. 2018 Aug 6;11:4583-4590. https://doi.org/10.2147/OTT.S141053 PMid:30122950 PMCid:PMC6084082
  56. Holderness BM, Malhotra S, Levy NB, Danilov AV. Brentuximab vedotin demonstrates activity in a patient with plasmablastic lymphoma arising from a background of chronic lymphocytic leukemia. J Clin Oncol. 2013 Apr 20;31(12):e197-9. https://doi.org/10.1200/JCO.2012.46.9593 PMid:23509308
  57. Luo B, Huang L, Gu Y, et al. Expression of exportin-1 in diffuse large B-cell lymphoma: immunohistochemistry and TCGA analyses. Int J Clin Exp Pathol 2018; 11: 5547-60.
  58. Culjkovic-Kraljacic B, Fernando TM, Marullo R, et al. Combinatorial targeting of nuclear export and translation of RNA inhibits aggressive B-cell lymphomas. Blood 2016; 127: 858-68. https://doi.org/10.1182/blood-2015-05-645069 PMid:26603836 PMCid:PMC4760090
  59. Kuruvilla J, Savona M, Baz R, et al. Selective inhibition of nuclear export with selinexor in patients with non-Hodgkin lymphoma. Blood 2017; 129: 3175-83. https://doi.org/10.1182/blood-2016-11-750174 PMid:28468797
  60. Laín S, Xirodimas D, Lane DP. Accumulating active p53 in the nucleus by inhibition of nuclear export: a novel strategy to promote the p53 tumor suppressor function. Exp Cell Res 1999; 253: 315-24 https://doi.org/10.1006/excr.1999.4672 PMid:10585254
  61. Chari A, Vogl DT, Gavriatopoulou M, et al. Oral selinexor dexamethasone for triple-class refractory multiple myeloma. N Engl J Med 2019; 381: 727-38 https://doi.org/10.1056/NEJMoa1903455 PMid:31433920
  62. Ben-Barouch S, Kuruvilla J. Selinexor (KTP-330) - a selective inhibitor of nuclear export (SINE): anti-tumor activity in diffuse large B-cell lymphoma (DLBCL). Expert Opin Investig Drugs 2019; 29: 15-21. https://doi.org/10.1080/13543784.2020.1706087 PMid:31847605
  63. Kasamon YL, Price LSL, Okusanya OO, Richardson NC, Li RJ, Ma L, et al. FDA Approval Summary: Selinexor for Relapsed or Refractory Diffuse Large B-Cell Lymphoma. Oncologist. 2021;26(10):879-86. https://doi.org/10.1002/onco.13859 PMid:34132444 PMCid:PMC8488790
  64. Podar K, Shah J, Chari A, Richardson PG, Jagannath S. Selinexor for the treatment of multiple myeloma. Expert Opin Pharmacother. 2020;21(4):399-408   PMid:31957504
  65. Yuhua F, Renjun B, Yonghua Y, Zhang T, Chen L. Selinexor in the Treatment of a Patient with Refractory Plasmablastic Lymphoma: A Case Reports. Ann Short Rep Clin Image. 2022; 3(3): 1028.
  66. Lin P, Owens R, Tricot G, Wilson CS. Flow cytometric immunophenotypic analysis of 306 cases of multiple myeloma. Am J Clin Pathol. 2004 Apr;121(4):482-8. https://doi.org/10.1309/74R4TB90BUWH27JX PMid:15080299
  67. de Weers M, Tai YT, van der Veer MS, Bakker JM, Vink T, Jacobs DC, Oomen LA, Peipp M, Valerius T, Slootstra JW, Mutis T, Bleeker WK, Anderson KC, Lokhorst HM, van de Winkel JG, Parren PW. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011 Feb 1;186(3):1840-8. https://doi.org/10.4049/jimmunol.1003032 PMid:21187443
  68. Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, Syed K, Liu K, van de Donk NW, Weiss BM, Ahmadi T, Lokhorst HM, Mutis T, Sasser AK. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016 Jul 21;128(3):384-94. https://doi.org/10.1182/blood-2015-12-687749 PMid:27222480 PMCid:PMC4957162
  69. Morabito F, Damle RN, Deaglio S et al. The CD38 ectoenzyme family: advances in basic science and clinical practice. Mol. Med. 2006; 12(11-12):342-4. https://doi.org/10.2119/2006-00110.Morabito PMid:17380202 PMCid:PMC1829202
  70. Patten PEM, Buggins AGS, Richards J et al. CD38 expression in chronic lymphocytic leukemia is regulated by the tumor microenvironment. Blood 2008; 111(10):5173-81. https://doi.org/10.1182/blood-2007-08-108605 PMid:18326821
  71. Konoplev S, Medeiros LJ, Bueso-Ramos CE et al. Immunophenotypic profile of lymphoplasmacytic lymphoma/Waldenström macroglobulinemia. Am. J. Clin. Pathol. 2005;124(3):414-20. https://doi.org/10.1309/3G1XDX0DVHBNVKB4 PMid:16191510
  72. Parry-Jones N, Matutes E, Morilla R et al. Cytogenetic abnormalities additional to t(11;14) correlate with clinical features in leukaemic presentation of mantle cell lymphoma, and may influence prognosis: A study of 60 cases by FISH. Br. J. Haematol. 2007; 137(2):117-124. https://doi.org/10.1111/j.1365-2141.2007.06526.x PMid:17391491
  73. Keyhani A, Huh YO, Jendiroba D et al. Increased CD38 expression is associated with favorable prognosis in adult acute leukemia. Leuk. Res. 2000; 24(2):153-9. https://doi.org/10.1016/S0145-2126(99)00147-2 PMid:10654451
  74. Marinov J, Koubek K, Starý J. Immunophenotypic significance of the "lymphoid" CD38 antigenin myeloid blood malignancies. Neoplasma 1993; 40(6):355-8.
  75. Suzuki R, Suzumiya J, Nakamura S et al. Aggressive natural killer-cell leukemia revisited:large granular lymphocyte leukemia of cytotoxic NK cells. Leukemia 2004; 18(4):763-70. https://doi.org/10.1038/sj.leu.2403262 PMid:14961041
  76. Wang L, Wang H, Li P et al. CD38 expression predicts poor prognosis and might be a potentialtherapy target in extranodal NK/T cell lymphoma, nasal type. Ann. Hematol. 2015;94(8):1381-8. https://doi.org/10.1007/s00277-015-2359-2 PMid:25865943
  77. Hari P, Raj R V, Olteanu H. Targeting CD38 in Refractory Extranodal Natural Killer Cell-T-CellLymphoma. N. Engl. J. Med. 2016; 375(15):1501-1502. https://doi.org/10.1056/NEJMc1605684 PMid:27732828
  78. Bride KL, Vincent TL, Im S-Y et al. Preclinical efficacy of daratumumab in T-cell acute lymphoblastic leukemia. Blood 2018; 131(9):995-999. https://doi.org/10.1182/blood-2017-07-794214 PMid:29305553 PMCid:PMC5833263
  79. Overdijk MB, Verploegen S, Bögels M et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. MAbs 2015; 7(2):311-21. https://doi.org/10.1080/19420862.2015.1007813 PMid:25760767 PMCid:PMC4622648
  80. Chikeka I, Grossman M, Deng C, Jacob AT, Husain S. Plasmablastic lymphoma in an HIV patient with cutaneous presentation: A case of remarkable remission in a typically refractory disease. JAAD Case Rep. 2020 Feb 12;6(3):161-165. https://doi.org/10.1016/j.jdcr.2019.11.007 PMid:32083160 PMCid:PMC7019043
  81. Marvyin K, Tjønnfjord EB, Breland UM, Tjønnfjord GE. Transformation to plasmablastic lymphoma in CLL upon ibrutinib treatment. BMJ Case Rep. 2020 Sep 29;13(9):e235816. https://doi.org/10.1136/bcr-2020-235816 PMid:32994268 PMCid:PMC7526319
  82. Roché P, Venton G, Berda-Haddad Y, Fritz S, Ivanov V, Mercier C, Colle J, Tichadou A, Fanciullino R, Lepidi H, Costello R, Farnault L. Could daratumumab induce the maturation of plasmablasts in Plasmablastic lymphoma?-Potential therapeutic applications. Eur J Haematol. 2021 Apr;106(4):589-592. https://doi.org/10.1111/ejh.13584 PMid:33469987
  83. Ricker E, Yun Kyoung Ryu, Jennifer E. Amengual, Daratumumab Plus Chemotherapy Induces Complete Responses in a Consecutive Series of Four Patients with Plasmablastic Lymphoma, Blood,Volume 138, Supplement 1,2021,Page 4573 https://doi.org/10.1182/blood-2021-150800
  84. Ramadas P, Williams M, Duggan DB. Plasmablastic Lymphoma or Plasmablastic Myeloma: A Case of Post-Transplant Lymphoproliferative Disorder. Case Rep Hematol. 2021 Sep 27;2021:4354941. https://doi.org/10.1155/2021/4354941 PMid:34616575 PMCid:PMC8490046
  85. Kathrotiya M, Radhakrishnan VS, Bhave SJ, Kumar J, Roychowdhury M, Arun I, Das J, Chandy M, Nair R. Relapsed plasmablastic lymphoma in a HIV-negative patient: Pushing the envelope. Clin Case Rep. 2020 Dec 20;9(2):873-877. https://doi.org/10.1002/ccr3.3673 PMid:33598263 PMCid:PMC7869339
  86. Ryu YK, Ricker EC, Soderquist CR, Francescone MA, Lipsky AH, Amengual JE. Targeting CD38 with Daratumumab Plus Chemotherapy for Patients with Advanced-Stage Plasmablastoid Large B-Cell Lymphoma. J Clin Med. 2022 Aug 22;11(16):4928. https://doi.org/10.3390/jcm11164928 PMid:36013165 PMCid:PMC9409851
  87. Bhatt P, Kloock C, Comenzo R. Relapsed/Refractory Multiple Myeloma: A Review of Available Therapies and Clinical Scenarios Encountered in Myeloma Relapse. Curr Oncol. 2023 Feb 15;30(2):2322-2347. https://doi.org/10.3390/curroncol30020179 PMid:36826140 PMCid:PMC9954856
  88. Pinto MP, Thorneloe NS, Brown MR, Stalons ML, Stoll KE, Holmes AR, Pathan M, Gonzales PA. The devolution of a mature plasma cell dyscrasia into a fatal plasmablastic lymphoma. J Case Rep Images Oncology 2023;9(2):7-14. https://doi.org/10.5348/100124Z10MP2023CR
  89. Dornan D, Bennett F, Chen Y et al (2009) Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkinlymphoma. Blood 114:2721-2729 https://doi.org/10.1182/blood-2009-02-205500 PMid:19633198
  90. Deeks ED (2019) Polatuzumab vedotin: first global approval. Drugs 79:1467-1475. https://doi.org/10.1007/s40265-019-01175-0 PMid:31352604 PMCid:PMC6794237
  91. Palanca-Wessels MCA, Czuczman M, Salles G et al (2015) Safety and activity of the anti-CD79B antibody-drug conjugate polatuzumab vedotin in relapsed or refractory B-cell non-Hodgkin lymphoma and chronic lymphocytic leukaemia: a phase 1 study. https://doi.org/10.1016/S1470-2045(15)70128-2 PMid:25925619
  92. Sehn L, Herrera A, Flowers C et al (2020) Polatuzumab vedotin in relapsed or refractory diffuse large B-cell lymphoma. J Clin Oncol 38(2):155-165 https://doi.org/10.1200/JCO.19.00172 PMid:31693429 PMCid:PMC7032881
  93. Northend M, Wilson W, Osborne W. et al.: Results of a United Kingdom real-world study of polatuzumab vedotin, bendamustine, and rituximab for relapsed/refractory DLBCL. Blood Adv. 2022 May 10;6(9):2920-2926. https://doi.org/10.1182/bloodadvances.2021005953 PMid:35020818 PMCid:PMC9092410
  94. Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. https://doi.org/10.3389/fphar.2017.00561 PMid:28878676 PMCid:PMC5572324
  95. Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887-3895. https://doi.org/10.1002/j.1460-2075.1992.tb05481.x PMid:1396582 PMCid:PMC556898
  96. Ishida, Y. PD-1: Its Discovery, Involvement in Cancer Immunotherapy, and Beyond. Cells 2020, 9, 1376. https://doi.org/10.3390/cells9061376 PMid:32492969 PMCid:PMC7349669
  97. Jalali, S.; Price-Troska, T.; Bothun, C.; Villasboas, J.; Kim, H.J.; Yang, Z.Z.; Novak, A.J.; Dong, H.; Ansell, S.M. Reverse signaling via PD-L1 supports malignant cell growth and survival in classical Hodgkin lymphoma. Blood Cancer J. 2019, 9, 22. https://doi.org/10.1038/s41408-019-0185-9 PMid:30783096 PMCid:PMC6381098
  98. Xu-Monette, Z.Y.; Zhou, J.; Young, K.H. PD-1 expression and clinical PD-1 blockade in B-cell lymphomas. Blood 2018, 131, 68-83. https://doi.org/10.1182/blood-2017-07-740993 PMid:29118007 PMCid:PMC5755041
  99. Garcia-Lacarte, M.; Grijalba, S.C.; Melchor, J.; Arnaiz-Leche, A.; Roa, S. The PD-1/PD-L1 Checkpoint in Normal Germinal Centers and Diffuse Large B-Cell Lymphomas. Cancers 2021, 13, 4683. https://doi.org/10.3390/cancers13184683 PMid:34572910 PMCid:PMC8471895
  100. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261-268. https://doi.org/10.1038/85330 PMid:11224527
  101. Xia, Y.; Medeiros, L.J.; Young, K.H. Immune checkpoint blockade: Releasing the brake towards hematological malignancies. Blood Rev. 2016, 30, 189-200. https://doi.org/10.1016/j.blre.2015.11.003 PMid:26699946
  102. Bardhan, K.; Anagnostou, T.; Boussiotis, V.A. The PD1:PD-L1/2 Pathway from Discovery to Clinical Implementation. Front.Immunol. 2016, 7, 550. https://doi.org/10.3389/fimmu.2016.00550
  103. Azuma, T.; Yao, S.; Zhu, G.; Flies, A.S.; Flies, S.J.; Chen, L. B7-H1 is a ubiquitous antiapoptotic receptor on cancer cells. Blood 2008,111, 3635-3643. https://doi.org/10.1182/blood-2007-11-123141 PMid:18223165 PMCid:PMC2275025
  104. Zhang, J.P.; Song, Z.; Wang, H.B.; Lang, L.; Yang, Y.Z.; Xiao, W.; Webster, D.E.; Wei, W.; Barta, S.K.; Kadin, M.E.; et al. A novel model of controlling PD-L1 expression in ALK(+) anaplastic large cell lymphoma revealed by CRISPR screening. Blood 2019, 134,171-185. https://doi.org/10.1182/blood.2019001043 PMid:31151983 PMCid:PMC6624970
  105. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.;Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014,515, 563-567. https://doi.org/10.1038/nature14011 PMid:25428504 PMCid:PMC4836193
  106. Xie, W.; Medeiros, L.J.; Li, S.; Yin, C.C.; Khoury, J.D.; Xu, J. PD-1/PD-L1 Pathway and Its Blockade in Patients with ClassicHodgkin Lymphoma and Non-Hodgkin Large-Cell Lymphomas. Curr. Hematol. Malig. Rep. 2020, 15, 372-381. https://doi.org/10.1007/s11899-020-00589-y PMid:32394185
  107. Ansell, S.M. PD-1 Blockade in Classic Hodgkin Lymphoma. JCO Oncol. Pract. 2021, 17, 72-73. [CrossRef] https://doi.org/10.1200/OP.20.01020 PMid:33439697
  108. Chen, B.J.; Chapuy, B.; Ouyang, J.; Sun, H.H.; Roemer, M.G.; Xu, M.L.; Yu, H.; Fletcher, C.D.; Freeman, G.J.; Shipp, M.A.; et al.PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin. Cancer Res.2013, 19, 3462-3473. https://doi.org/10.1158/1078-0432.CCR-13-0855 PMid:23674495 PMCid:PMC4102335
  109. Kiyasu, J.; Miyoshi, H.; Hirata, A.; Arakawa, F.; Ichikawa, A.; Niino, D.; Sugita, Y.; Yufu, Y.; Choi, I.; Abe, Y.; et al. Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. Blood2015, 126, 2193-2201 https://doi.org/10.1182/blood-2015-02-629600 PMid:26239088 PMCid:PMC4635115
  110. Granai M, Mundo L, Akarca AU, Siciliano MC, Rizvi H, Mancini V, Onyango N, Nyagol J, Abinya NO, Maha I, Margielewska S, Wi W, Bibas M, Piccaluga PP, Quintanilla-Martinez L, Fend F, Lazzi S, Leoncini L, Marafioti T. Immune landscape in Burkitt lymphoma reveals M2-macrophage polarization and correlation between PD-L1 expression and non-canonical EBV latency program. Infect Agent Cancer. 2020 May 6;15:28. https://doi.org/10.1186/s13027-020-00292-w PMid:32391073 PMCid:PMC7201729
  111. Xing, W.; Dresser, K.; Zhang, R.; Evens, A.M.; Yu, H.; Woda, B.A.; Chen, B.J. PD-L1 expression in EBV-negative diffuse large B-cell lymphoma: Clinicopathologic features and prognostic implications. Oncotarget 2016, 7, 59976-59986. https://doi.org/10.18632/oncotarget.11045 PMid:27527850 PMCid:PMC5312363
  112. Laurent C, Fabiani B, Do C, Tchernonog E, Cartron G, Gravelle P, Amara N, Malot S, Palisoc MM, Copie-Bergman C, Glehen AT, Copin MC, Brousset P, Pittaluga S, Jaffe ES, Coppo P. Immune-checkpoint expression in Epstein-Barr virus positive and negative plasmablastic lymphoma: a clinical and pathological study in 82 patients. Haematologica. 2016 Aug;101(8):976-84. https://doi.org/10.3324/haematol.2016.141978 PMid:27175027 PMCid:PMC4967577
  113. Gravelle P, Péricart S, Tosolini M, Fabiani B, Coppo P, Amara N, Traverse-Gléhen A, Van Acker N, Brousset P, Fournie JJ, Laurent C. EBV infection determines the immune hallmarks of plasmablastic lymphoma. Oncoimmunology. 2018 Jul 30;7(10):e1486950. https://doi.org/10.1080/2162402X.2018.1486950 PMid:30288350 PMCid:PMC6169584
  114. Damlaj M., Alzayed M., Alahmari B., Alhejazi A., Alaskar A., Alzahrani M. Therapeutic potential of checkpoint inhibitors in refractory plasmablastic lymphoma. Clinical Lymphoma Myeloma and Leukemia . 2019;19(10):e559-e563. https://doi.org/10.1016/j.clml.2019.06.008 PMid:31377210
  115. Cheng L., Song Q., Liu M., et al. Case report: successful management of a refractory plasmablastic lymphoma patient with tislelizumab and lenalidomide. Frontiers in Immunology . 2021;12 https://doi.org/10.3389/fimmu.2021.702593 PMid:34322131 PMCid:PMC8312258
  116. Castillo JJ, Lamacchia J, Silver J, Flynn CA, Sarosiek S. Complete response to pembrolizumab and radiation in a patient with HIV-negative, EBV-positive plasmablastic lymphoma. Am J Hematol. 2021 Oct 1;96(10):E390-E392. https://doi.org/10.1002/ajh.26291 PMCid:PMC8252623
  117. Schuster, S.J. , Svoboda, J. , Chong, E.A. , Nasta, S.D. , Mato, A.R. , Anak, O., Brogdon, J.L., Pruteanu-Malinici, I., Bhoj, V., Landsburg, D. , Wasik, M., Levine, B.L., Lacey, S.F., Melenhorst, J.J., Porter, D.L. & June, C.H. (2017) Chimeric antigen receptor T cells in refractory B-cell lymphomas. The New England Journal of Medicine, 377, 2545-2554. https://doi.org/10.1056/NEJMoa1708566 PMid:29226764 PMCid:PMC5788566
  118. Abramson, J.S. & Chen, Y.B. (2017) More on anti-CD19 CAR T cells in CNS diffuse large-B-cell lymphoma. The New England Journal of Medicine, 377, 2102. https://doi.org/10.1056/NEJMc1704610
  119. Testa U, Leone G, Pelosi E, Castelli G, Hohaus S. CAR-T Cell Therapy in Large B Cell Lymphoma. Mediterr J Hematol Infect Dis. 2023 Nov 1;15(1):e2023066. https://doi.org/10.4084/MJHID.2023.066 PMid:38028399 PMCid:PMC10631715
  120. Parker C, Liu FF, Deger KA, Franco-Villalobos C, Proskorovsky I, Keating SJ, Sorensen S. Cost-Effectiveness of Lisocabtagene Maraleucel Versus Axicabtagene Ciloleucel and Tisagenlecleucel in the Third-Line or Later Treatment Setting for Relapsed or Refractory Large B-cell Lymphoma in the United States. Adv Ther. 2023 May;40(5):2355-2374. https://doi.org/10.1007/s12325-023-02444-x PMid:36947328 PMCid:PMC10129927
  121. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Reagan PM, Miklos DB, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, Crump M, Kuruvilla J, Van Den Neste E, Farooq U, Navale L, DePuy V, Kim JJ, Gisselbrecht C. Comparison of 2-year outcomes with CAR T cells (ZUMA-1) vs salvage chemotherapy in refractory large B-cell lymphoma. Blood Adv. 2021 Oct 26;5(20):4149-4155. https://doi.org/10.1182/bloodadvances.2020003848 PMid:34478487 PMCid:PMC8945634
  122. Maziarz RT, Zhang J, Yang H, Chai X, Yuan C, Schwartz E, Jakovach M, Martinez-Prieto M, Agarwal A, Degtyarev E, et al. Indirect comparison of tisagenlecleucel and historical treatments for relapsed/refractory diffuse large B-cell lymphoma. Blood Adv 2022; 6: 2536-2542. https://doi.org/10.1182/bloodadvances.2021006280 PMid:35030634 PMCid:PMC9043930
  123. Bishop MR, Dickinson M, Purtill D, Barba P, Santoro A, Hamad N, Kato K, Sureda A, Greil R, Thieblemont C. Second-line tisagenlecleucel or standard care in aggressive B-cell lymphoma. N Engl J Med 2022, 386(7): 629-639. https://doi.org/10.1056/NEJMoa2116596 PMid:34904798
  124. Manson G, Cartron G, Beauvais D, Roulin L, Gros FX, et al. A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma. Nature Med 2022; 28: 2145- https://doi.org/10.1038/s41591-022-01969-y PMid:36138152 PMCid:PMC9556323
  125. Raychaudhuri R, Qualtieri J, Garfall AL. Axicabtagene ciloleucel for CD19+ plasmablastic lymphoma. Am J Hematol. 2020 Jan;95(1):E28-E30. https://doi.org/10.1002/ajh.25682 PMid:31725923
  126. Al-Malki MM, Castillo JJ, Sloan JM, Re A. Hematopoietic cell transplantation for plasmablastic lymphoma: a review. Biol Blood Marrow Transplant. 2014 Dec;20(12):1877-84. https://doi.org/10.1016/j.bbmt.2014.06.009 PMid:24946718
  127. Cattaneo C, Finel H, McQuaker G, Vandenberghe E, Rossi G, Dreger P. Autologous hematopoietic stem cell transplantation for plasmablastic lymphoma: the European Society for Blood and Marrow Transplantation experience. Biol Blood Marrow Transplant. 2015 Jun;21(6):1146-7. https://doi.org/10.1016/j.bbmt.2015.03.008 PMid:25783635
  128. Balsalobre P, Diez-Martin J, Re A, et al. Autologous stem cell transplantation in patients with HIV related lymphoma. J Clin Oncol. 2009; 27:2192-2198 https://doi.org/10.1200/JCO.2008.18.2683 PMid:19332732
  129. Re A, Michieli M, Allione B, et al. Early consolidation with high dose therapy and autologous stem cell transplantation in HIV-associated Non Hodgkin Lymphoma at high risk (aa-IPI 2-3), an interim report of a multicenter phase II trial. Blood. 2012;120. Abstract 731. https://doi.org/10.1182/blood.V120.21.3124.3124
  130. Cattaneo C, Re A, Ungari M, et al. Plasmablastic lymphoma among HIVpositive patients: results of a single centre's experience. Leuk Lymphoma. 2014 Apr 9 [Epub ahead of print]. https://doi.org/10.3109/10428194.2014.911867 PMid:24712980
  131. Broccoli A, Nanni L, Stefoni V, Agostinelli C, Argnani L, Cavo M, Zinzani PL. A patient with plasmablastic lymphoma achieving long-term complete remission after thalidomide-dexamethasone induction and double autologous stem cell transplantation: a case report. BMC Cancer. 2018 Jun 8;18(1):645. https://doi.org/10.1186/s12885-018-4561-9 PMid:29879938 PMCid:PMC5992724
  132. M. Hamadani, S.M. Devine.Reduced-intensity conditioning allogeneic stem cell transplantation in HIV patients with hematologic malignancies: yes, we can. Blood,114 (2009), pp. 2564-2566 https://doi.org/10.1182/blood-2009-06-229666 PMid:19762505
  133. Rong C, Sheng L, Wu A, Sun Y, Ouyang G. Allogeneic hematopoietic stem cell transplantation in a patient with HIV-negative recurrent plasmablastic lymphoma: A case report. Medicine (Baltimore). 2021 Feb 19;100(7):e24498. https://doi.org/10.1097/MD.0000000000024498 PMid:33607779 PMCid:PMC7899902
  134. Illidge T, Specht L, Yahalom J, et al. Modern Radiation Therapy for Nodal Non-Hodgkin Lymphoma-Target Definition and Dose Guidelines From the International Lymphoma Radiation Oncology Group. International Journal of Radiation Oncology*Biology*Physics 2014;89(1):49-58 https://doi.org/10.1016/j.ijrobp.2014.01.006 PMid:24725689
  1. Ng AK, Yahalom J, Goda JS, et al. Role of Radiation Therapy in Patients With Relapsed/Refractory Diffuse Large B-Cell Lymphoma: Guidelines from the International Lymphoma Radiation Oncology Group. International Journal of Radiation Oncology*Biology*Physics 2018;100(3):652-69. https://doi.org/10.1016/j.ijrobp.2017.12.005 PMid:29413279
  2. Shi Z, Das S, Okwan-Duodu D, et al. Patterns of failure in advanced stage diffuse large B-cell lymphoma patients after complete response to R-CHOP immunochemotherapy and the emerging role of consolidative radiation therapy. Int J Radiat Oncol Biol Phys. 2013;86(3):569-577. https://doi.org/10.1016/j.ijrobp.2013.02.007 PMid:23540349
  3. Phan J, Mazloom A, Medeiros LJ, et al. Benefit of consolidative radiation therapy in patients with diffuse large B-cell lymphoma treated with R-CHOP chemotherapy. J Clin Oncol. 2010;28(27):4170-4176. https://doi.org/10.1200/JCO.2009.27.3441 PMid:20713859
  4. Horning SJ, Weller E, Kim K, et al. Chemotherapy with or without radiotherapy in limited-stage diffuse aggressive non-Hodgkin's lymphoma: Eastern Cooperative Oncology Group study 1484. J Clin Oncol. 2004;22(15):3032-3038. https://doi.org/10.1200/JCO.2004.06.088 PMid:15210738
  5. Miller TP, Dahlberg S, Cassady JR, et al. Chemotherapy alone compared with chemotherapy plus radiotherapy for localized intermediate- and high-grade non-Hodgkin's lymphoma. N Engl J Med. 1998;339(1):21-26. https://doi.org/10.1056/NEJM199807023390104 PMid:9647875
  6. Held G, Murawski N, Ziepert M, et al. Role of radiotherapy to bulky disease in elderly patients with aggressive B-cell lymphoma. J Clin Oncol. 2014;32(11):1112-1118. https://doi.org/10.1200/JCO.2013.51.4505 PMid:24493716
  7. Pfreundschuh M, Murawski N, Ziepert M, et al. Radiotherapy (RT) to bulky (B) and extralymphatic (E) disease in combination with 6xR-CHOP-14 or R-CHOP-21 in young good-prognosis DLBCL patients: results of the 2x2 randomized UNFOLDER trial of the DSHNHL/GLA. J Clin Oncol. 2018;36(15_suppl):7574-7574. https://doi.org/10.1200/JCO.2018.36.15_suppl.7574
  8. Held G, Zeynalova S, Murawski N, et al. Impact of rituximab and radiotherapy on outcome of patients with aggressive B-cell lymphoma and skeletal involvement. J Clin Oncol. 2013;31(32):4115-4122. https://doi.org/10.1200/JCO.2012.48.0467 PMid:24062391
  9. Ng AK, Yahalom J, Goda JS, et al. Role of radiation therapy in patients with relapsed/refractory diffuse large B-cell lymphoma: guidelines from the International Lymphoma Radiation Oncology Group. Int J Radiat Oncol Biol Phys. 2018;100(3):652-669. https://doi.org/10.1016/j.ijrobp.2017.12.005 PMid:29413279
  10. Wright CM, Koroulakis AI, Baron JA, et al. Palliative radiotherapy for diffuse large B-cell lymphoma. Clin Lymphoma Myeloma Leuk. 2021;21(10):650-658. https://doi.org/10.1016/j.clml.2021.05.007 PMid:34127417
  11. Liu M, Liu B, Liu B, Wang Q, Ding L, Xia C, Dong L. Human immunodeficiency virus-negative plasmablastic lymphoma: a comprehensive analysis of 114 cases. Oncol Rep. 2015 Apr;33(4):1615-20. https://doi.org/10.3892/or.2015.3808  PMid:25695332 PMCid:PMC4358079
  12. Tchernonog E, Faurie P, Coppo P, Monjanel H, Bonnet A, Algarte Génin M, Mercier M, Dupuis J, Bijou F, Herbaux C, Delmer A, Fabiani B, Besson C, Le Gouill S, Gyan E, Laurent C, Ghesquieres H, Cartron G. Clinical characteristics and prognostic factors of plasmablastic lymphoma patients: analysis of 135 patients from the LYSA group. Ann Oncol. 2017 Apr 1;28(4):843-848. https://doi.org/10.1093/annonc/mdw684 PMid:28031174
  13. Hess BT, Giri A, Park Y, Patel KK, Link BK, Nowakowski GS, Maliske SM, Fortin S, Chavez JC, Saeed H, Hill BT, Mejia Garcia AV, Maddocks KJ, Hanel W, Wagner-Johnston ND, Messmer MR, Kahl BS, Watkins M, Alderuccio JP, Lossos IS, Nathan S, Orellana-Noia VM, Portell CA, Landsburg DJ, Ayers EC, Castillo JJ. Outcomes of patients with limited-stage plasmablastic lymphoma: A multi-institutional retrospective study. Am J Hematol. 2023 Feb;98(2):300-308. https://doi.org/10.1002/ajh.26784 PMid:36588409 PMCid:PMC10107934
  14. Gleeson M, Counsell N, Cunningham D, et al. Central nervous system relapse of diffuse large B-cell lymphoma in the rituximab era: results of the UK NCRI R-CHOP-14 versus 21 trial. Ann Oncol. 2017;28(10):2511-2516. https://doi.org/10.1093/annonc/mdx353 PMid:28961838 PMCid:PMC5834096
  15. Ghose A, Elias HK, Guha G, Yellu M, Kundu R, Latif T. Influence of rituximab on central nervous system relapse in diffuse large B-cell lymphoma and role of prophylaxis-a systematic review of prospective studies. Clin Lymphoma Myeloma Leuk. 2015;15(8):451-457. https://doi.org/10.1016/j.clml.2015.02.026 PMid:25816933
  16. Klanova M, Sehn LH, Bence-Bruckler I, et al. Integration of cell of origin into the clinical CNS International Prognostic Index improves CNS relapse prediction in DLBCL. Blood. 2019;133(9):919-926. https://doi.org/10.1182/blood-2018-07-862862 PMid:30617197 PMCid:PMC6396175
  17. Schmitz N, Zeynalova S, Nickelsen M, et al. CNS International Prognostic Index: a risk model for CNS relapse in patients with diffuse large B-cell lymphoma treated with R-CHOP. J Clin Oncol. 2016;34(26):3150-3156. https://doi.org/10.1200/JCO.2015.65.6520 PMid:27382100
  18. Savage KJ. Secondary CNS relapse in diffuse large B-cell lymphoma: defining high-risk patients and optimization of prophylaxis strategies. Hematology Am Soc Hematol Educ Program. 2017;2017(1):578-586. https://doi.org/10.1182/asheducation-2017.1.578 PMid:29222307 PMCid:PMC6142549
  19. Kridel R, Telio D, Villa D, et al. Diffuse large B-cell lymphoma with testicular involvement: outcome and risk of CNS relapse in the rituximab era. Br JHaematol. 2017;176(2):210-221. https://doi.org/10.1111/bjh.14392 PMid:27739058
  20. Hu S, Song Y, Li Y, et al. Primary breast diffuse large B cell lymphoma in the rituximab era: outcomes of a multicenter retrospective study by the Lymphoma and Leukemia Committee of Chinese Geriatric Oncology Society (LLC-CGOS). Paper presented at: 58th Annual Meeting of the American Society of Hematology; 3-6 December 2016; San Diego, CA.
  21. McKay P, Wilson MR, Chaganti S, et al; British Society of Haematology. The prevention of central nervous system relapse in diffuse large B-cell lymphoma: a British Society for Haematology Good Practice Paper. Br J Haematol. 2020;190(5):708-714. https://doi.org/10.1111/bjh.16866 PMid:32433789
  22. El-Galaly TC, Villa D, Michaelsen TY, et al. The number of extranodal sites assessed by PET/CT scan is a powerful predictor of CNS relapse for patients with diffuse large B-cell lymphoma: an international multicenter study of 1532 patients treated with chemoimmunotherapy. Eur J Cancer.2017;75(April):195-203. https://doi.org/10.1016/j.ejca.2016.12.029 PMid:28237865
  23. Savage KJ, Slack GW, Mottok A, et al. Impact of dual expression of MYC and BCL2 by immunohistochemistry on the risk of CNS relapse in DLBCL.Blood. 2016;127(18):2182-2188 https://doi.org/10.1182/blood-2015-10-676700 PMid:26834242
  24. Wilson WH, Bromberg JE, Stetler-Stevenson M, et al. Detection and outcome of occult leptomeningeal disease in diffuse large B-cell lymphoma and Burkitt lymphoma. Haematologica. 2014;99(7):1228-1235. https://doi.org/10.3324/haematol.2013.101741 PMid:24727817 PMCid:PMC4077085
  25. Eyre TA, Djebbari F, Kirkwood AA, Collins GP. Efficacy of central nervous system prophylaxis with stand-alone intrathecal chemotherapy in diffuse large B-cell lymphoma patients treated with anthracycline-based chemotherapy in the rituximab era: a systematic review. Haematologica.2020;105(7):1914-1924. https://doi.org/10.3324/haematol.2019.229948 PMid:31488560 PMCid:PMC7327624
  26. Eyre TA, Kirkwood AA, Wolf J, et al. Stand-alone intrathecal central nervous system (CNS) prophylaxis provide unclear benefit in reducing CNS relapse risk in elderly DLBCL patients treated with R-CHOP and is associated increased infection-related toxicity. Br J Haematol. 2019;187(2):185-194 https://doi.org/10.1111/bjh.16070 PMid:31222719
  27. Barta SK, Joshi J, Mounier N, et al. Central nervous system involvement in AIDS-related lymphomas. Br J Haematol 2016;173:857-66. https://doi.org/10.1111/bjh.13998 PMid:27062389 PMCid:PMC4900917
  28. Hoffmann C, Wolf E, Fatkenheuer G, Buhk T, Stoehr A, Plettenberg A, et al.Response to highly active antiretroviral therapy strongly predicts outcome in patients with AIDS-related lymphoma. AIDS. 2003;17:1521-9. https://doi.org/10.1097/00002030-200307040-00013 PMid:12824790
  29. Lim ST, Karim R, Nathwani BN, Tulpule A, Espina B, Levine AM. AIDS-related Burkitt's lymphoma versus diffuse large-cell lymphoma in the pre-highly active antiretroviral therapy (HAART) and HAART eras: significant differences in survival with standard chemotherapy. J Clin Oncol. 2005;23:4430-8. https://doi.org/10.1200/JCO.2005.11.973 PMid:15883411
  30. Navarro JT, Ribera JM, Oriol A, Vaquero M, Romeu J, Batlle M, et al. Influence of highly active anti-retroviral therapy on response to treatment and survival in patients with acquired immunodeficiency syndrome-related non-Hodgkin's lymphoma treated with cyclophosphamide, hydroxydoxorubicin, vincristine and prednisone. Br J Haematol. 2001;112:909-15. https://doi.org/10.1046/j.1365-2141.2001.02656.x PMid:11298585
  31. Vaccher E, Spina M, di Gennaro G, Talamini R, Nasti G, Schioppa O, et al Concomitant cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy plus highly active antiretroviral therapy in patients with human immunodeficiency virus-related, non-Hodgkin lymphoma. Cancer. 2001;91:155-63. https://doi.org/10.1002/1097-0142(20010101)91:1<155::AID-CNCR20>3.0.CO;2-B PMid:11148572
  32. Torres HA, Rallapalli V, Saxena A, Granwehr BP, Viola GM, Ariza-Heredia E, et al Efficacy and safety of antiretrovirals in HIV-infected patients with cancer. Clin Microbiol Infect. 2014;20:O672-9. https://doi.org/10.1111/1469-0691.12589 PMid:24529214
  33. Gonzalez-Garcia JJ, Mahillo B, Hernandez S, Pacheco R, Diz S, Garcia P, et al. [Prevalences of hepatitis virus coinfection and indications for chronic hepatitis C virus treatment and liver transplantation in Spanish HIV-infected patients.The GESIDA 29/02 and FIPSE 12185/01 Multicenter Study]. Enferm Infecc Microbiol Clin. 2005;23:340-8. https://doi.org/10.1157/13076173 PMid:15970166
  34. Konopnicki D, Mocroft A, de Wit S, Antunes F, Ledergerber B, Katlama C, et al. Hepatitis B and HIV: prevalence, AIDS progression, response to highly active antiretroviral therapy and increased mortality in the EuroSIDA cohort. AIDS.2005;19:593-601. https://doi.org/10.1097/01.aids.0000163936.99401.fe PMid:15802978
  35. Berenguer J, Rivero A, Jarrin I, Nunez MJ, Vivancos MJ, Crespo M, et al. Human immunodeficiency virus/hepatitis C virus coinfection in Spain: prevalence and patient characteristics. Open Forum Infect Dis. 2016;3:ofw059.
  36. Stebbing J, Atkins M, Nelson M, Rajpopat S, Newsom-Davis T, Gazzard B,et al. Hepatitis B reactivation during combination chemotherapy for AIDSrelated lymphoma is uncommon and does not adversely affect outcome. Blood.2004;103:2431-2. https://doi.org/10.1182/blood-2003-12-4222 PMid:14998920
  37. Stroffolini T, Andriani A, Bibas M, Barlatani A. Successful treatment with lamivudine for reactivated hepatitis B infection following chemotherapy for non-Hodgkin's lymphoma. Ann Hematol. 2002 Jan;81(1):48-9. https://doi.org/10.1007/s00277-001-0393-8 PMid:11807636
  38. Lalazar G, Rund D, Shouval D. Screening, prevention and treatment of viral hepatitis B reactivation in patients with haematological malignancies. Br JHaematol. 2007;136:699-712. https://doi.org/10.1111/j.1365-2141.2006.06465.x PMid:17338776
  39. Evens AM, Jovanovic BD, Su YC, Raisch DW, Ganger D, Belknap SM, et al.Rituximab-associated hepatitis B virus (HBV) reactivation in lymphoproliferative diseases: meta-analysis and examination of FDA safety reports. Ann Oncol.2011;22:1170-80. https://doi.org/10.1093/annonc/mdq583 PMid:21115603 PMCid:PMC3082161
  40. Price H, Dunn D, Pillay D, Bani-Sadr F, de Vries-Sluijs T, Jain MK, et al. Suppression of HBV by tenofovir in HBV/HIV coinfected patients: a systematic review and meta-analysis. PLOS ONE. 2013;8:e68152. https://doi.org/10.1371/journal.pone.0068152 PMid:23874527 PMCid:PMC3707972
  41. Panel de expertos de GeSIDA y Plan Nacional sobre el Sida. Documento de consenso de GeSIDA/Plan Nacional sobre el Sida respect al tratamiento antirretroviral en adultos infecta- dos por el virus dela inmunodeficiencia humana. Available from: http://gesida-seimc.org/wp-content/uploads/2017/02/gesida-guiasclinicas-2017-TAR.pdf
  42. Buti M, Gane E, Seto WK, Chan HL, Chuang WL, Stepanova T, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate for the treatment of patients with HBeAg-negative chronic hepatitis B virus infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet Gastroenterol Hepatol. 2016;1:196-206. https://doi.org/10.1016/S2468-1253(16)30107-8 PMid:28404092
  43. Chan HL, Fung S, Seto WK, Chuang WL, Chen CY, Kim HJ, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate for the treatment of HBeAgpositive chronic hepatitis B virus infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet Gastroenterol Hepatol. 2016;1:185-95. https://doi.org/10.1016/S2468-1253(16)30024-3 PMid:28404091
  44. Alwan F, He A, Montoto S, Kassam S, Mee M, Burns F, et al. Adding rituximab to CODOX-M/IVAC chemotherapy in the treatment of HIV-associated Burkittlymphoma is safe when used with concurrent combination antiretroviral therapy.AIDS. 2015;29:903-10. https://doi.org/10.1097/QAD.0000000000000623 PMid:25730506
  45. Bower M, McCall-Peat N, Ryan N, Davies L, Young AM, Gupta S, et al. Protease inhibitors potentiate chemotherapy-induced neutropenia. Blood.2004;104:2943-6. https://doi.org/10.1182/blood-2004-05-1747 PMid:15238428
  46. Barta SK, Xue X, Wang D, Tamari R, Lee JY, Mounier N, et al. Treatment factors affecting outcomes in HIV-associated non-Hodgkin lymphomas: a pooledanalysis of 1546 patients. Blood. 2013;122:3251-62. https://doi.org/10.1182/blood-2013-04-498964 PMid:24014242 PMCid:PMC3821722
  47. Rudek MA, Flexner C, Ambinder RF. Use of antineoplastic agents in patients with cancer who have HIV/AIDS. Lancet Oncol. 2011;12:905-12. https://doi.org/10.1016/S1470-2045(11)70056-0 PMid:21570912
  48. Tourret J, Deray G, Isnard-Bagnis C. Tenofovir effect on the kidneys of HIVinfected patients: a double-edged sword? J Am Soc Nephrol. 2013;24:1519-27. https://doi.org/10.1681/ASN.2012080857 PMid:24052632 PMCid:PMC3785270
  49. Antela A, Aguiar C, Compston J, Hendry BM, Boffito M, Mallon P, et al. The role of tenofovir alafenamide in future HIV management. HIV Med. 2016;17 Suppl. 2:4-16. https://doi.org/10.1111/hiv.12401 PMid:26952360
  50. Goldman SC, Holcenberg JS, Finklestein JZ, Hutchinson R, Kreissman S, Johnson FL, et al. A randomized comparison between rasburicase and allopurinol in children with lymphoma or leukemia at high risk for tumor lysis. Blood.2001;97:2998-3003. https://doi.org/10.1182/blood.V97.10.2998 PMid:11342423
  51. Pui CH, Mahmoud HH, Wiley JM, Woods GM, Leverger G, Camitta B, et al. Recombinant urate oxidase for the prophylaxis or treatment of hyperuricemia in patients With leukemia or lymphoma. J Clin Oncol. 2001;19:697-704. https://doi.org/10.1200/JCO.2001.19.3.697 PMid:11157020
  52. Smith TJ, Khatcheressian J, Lyman GH, Ozer H, Armitage JO, Balducci L, et al. 2006 update of recommendations for the use of white blood cell growthfactors: an evidence-based clinical practice guideline. J Clin Oncol. 2006;24:3187-205. https://doi.org/10.1200/JCO.2006.06.4451 PMid:16682719
  53. Tomblyn M, Chiller T, Einsele H, Gress R, Sepkowitz K, Storek J, et al. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant.2009;15:1143-238. https://doi.org/10.1016/j.bbmt.2009.06.019 PMid:19747629 PMCid:PMC3103296
  54. Freifeld AG, Bow EJ, Sepkowitz KA, Boeckh MJ, Ito JI, Mullen CA, et al. Clinical practice guideline for the use of antimicrobial agents in neutropenic patientswith cancer: 2010 update by the Infectious Diseases Society of America. ClinInfect Dis. 2011;52:e56-93. https://doi.org/10.1093/cid/cir073 PMid:21258094
  55. Flowers CR, Seidenfeld J, Bow EJ, Karten C, Gleason C, Hawley DK, et al. Antimicrobial prophylaxis and outpatient management of fever and neutropenia in adults treated for malignancy: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2013;31:794-810. https://doi.org/10.1200/JCO.2012.45.8661 PMid:23319691
  56. Beck CR, McKenzie BC, Hashim AB, Harris RC, University of Nottingham Influenza and the ImmunoCompromised Study Group, Nguyen-Van-Tam JS. Influenza vaccination for immunocompromised patients: systematic review and meta-analysis by etiology. J Infect Dis. 2012;206:1250-9. https://doi.org/10.1093/infdis/jis487 PMid:22904335
  57. Rubin LG, Levin MJ, Ljungman P, Davies EG, Avery R, Tomblyn M, et al. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis. 2014;58:309-18 https://doi.org/10.1093/cid/cit816 PMid:24421306
  58. Mondi A, Lorenzini P, Castilletti C, .:INMI Recovery study group. Risk and predictive factors of prolonged viral RNA shedding in upper respiratory specimens in a large cohort of COVID-19 patients admitted to an Italian reference hospital. Int J Infect Dis. 2021 Apr;105:532-539.
  59. Buske C, Dreyling M, Alvarez-Larrán A, et al.: Managing hematological cancer patients during the COVID-19 pandemic: an ESMO-EHA Interdisciplinary Expert Consensus. ESMO Open. 2022 Apr;7(2):100403. https://doi.org/10.1016/j.esmoop.2022.100403 PMid:35272130 PMCid:PMC8795783