Abstract
The therapeutic options available for patients with multiple myeloma have greatly expanded over the past decade and incorporating these novel agents into routine clinical practice has significantly improved outcomes. The next generation of therapeutics is available for relapsed and refractory patients either as standard of care or in clinical trial, and these drugs represent a generational paradigm shift. Patients now have access to a multitude of novel immunotherapeutics, including monoclonal antibodies, an antibody–drug conjugate, chimeric antigen receptor T-cells (CAR-T), and bispecific T-cell redirecting antibodies, and novel oral therapies including selinexor (selective inhibitor of nuclear export) and venetoclax (bcl-2 inhibitor). While these drugs have the potential to be highly efficacious in certain subsets of patients when used as single agents or in combination regimens, they are each associated with unique toxicity profiles. It is imperative to understand these potential adverse events to ensure patient safety. Appropriate supportive care management is paramount to maximize drug exposure and therapeutic efficacy. The following review focuses its discussion on drugs and combination regimens that are currently FDA-approved and those that continue to be investigated in clinical trials, highlights the clinically relevant toxicity profiles for each of the different agents, and provides practical considerations for the treatment team.
Keywords: BCMA, belantamab mafodotin, CAR-T, ciltacaptagene autoleucel, daratumumab, elotuzumab, idecabtagene vicleucel, isatuximab, monoclonal antibody, relapsed and refractory multiple myeloma, selinexor, T-cell redirecting bispecific antibody, venetoclax
Introduction
Multiple myeloma (MM) is a hematologic cancer characterized by malignant transformation of plasma cells, a terminally differentiated B-cell. In 2021, there was an estimated ~34,000 new cases and ⩾12,000 deaths in the United States. 1 In most, MM is associated with production of a monoclonal protein and end-organ damage that may include renal dysfunction, anemia, bone lytic lesions, and hypercalcemia. Despite dramatic improvements in treatment options, the disease remains incurable, but patients treated with contemporary approaches have an improved overall survival over 10 years. 2
Foundational shifts in the MM treatment landscape have occurred over the last 20 years. The clinical development and FDA approval of immunomodulatory drugs (IMiDs) and proteosome inhibitors (PIs) in the early 2000s, along with the recent emergence of monoclonal antibodies (mAbs), were fundamental breakthroughs responsible for improving survival outcomes (Table 1). Other immunotherapeutics are now changing our approach to treatment. Antibody–drug conjugates (ADC), chimeric antigen receptor T-cells (CAR-T) and T-cell redirecting bispecific antibodies, alongside drugs with novel antitumor mechanisms, such as selinexor and venetoclax, are leading the next revolution and shifting us away from conventional chemotherapy toward effective, and in some cases, better tolerated interventions. However, these next-generation therapeutics are not without toxicities. Each drug class is associated with unique side effect profiles. Understanding these toxicities and initiating appropriate supportive and preventive strategies is critical for maximum drug exposure, improved response, and quality of life for our patients. In the present review, we highlight specific drugs within each of these novel drug classes to provide an overview of relevant side effect profiles and discuss practical management strategies.
Table 1.
Selected FDA approved therapies for the treatment of newly diagnosed and relapsed/refractory multiple myeloma from 2015-2022.
Drug +/− combination | Approval year | Indication |
---|---|---|
DARATUMUMAB (IV) | 2015 | RRMM at least three prior lines including IMiD and PI |
DARATUMUMAB (IV) + Rd | 2016 | RRMM at least one prior line |
DARATUMUMAB (IV) + Vd | 2016 | RRMM at least one prior line |
DARATUMUMAB (IV) + Pd | 2017 | RRMM at least two prior lines including PI and lenalidomide |
ELOTUZUMAB + Rd | 2015 | RRMM one to three prior lines |
ELOTUZUMAB + Pd | 2018 | RRMM at least two prior lines including PI and lenalidomide |
DARATUMUMAB + VTd | 2019 | Transplant eligible NDMM |
DARATUMUMAB + Rd | 2019 | Transplant ineligible NDMM |
SELINEXOR + Dex | 2019 | RRMM at least four prior lines including IMiD (2), PI (2), and aCD38 mAb |
SELINEXOR + Vd | 2020 | RRMM at least one prior line |
DARATUMUMAB (IV) + Kd | 2020 | RRMM one to three prior lines |
DARATUMUMAB (SC) | 2020 | RRMM at least three prior lines including IMiD and PI or double-refractory |
DARATUMUMAB (SC) + Rd | 2020 | RRMM at least one prior line |
DARATUMUMAB (SC) + Vd | 2020 | RRMM at least one prior line |
BELANTAMAB MAFODOTIN | 2020 | RRMM at least four prior lines including IMiD, PI, and aCD38 mAb |
ISATUXIMAB + Pd | 2020 | RRMM at least two prior lines including PI and lenalidomide |
ISATUXIMAB + Kd | 2021 | RRMM one to three prior lines |
IDECAPTAGENE VICLEUCEL | 2021 | RRMM at least four prior lines including IMiD, PI, and aCD38 mAb |
DARATUMUMAB (SC) + Pd | 2021 | RRMM at least one prior line including IMiD and PI |
DARATUMUMAB (SC) + Kd | 2021 | RRMM at least one prior line including PI and lenalidomide |
CILTACABTAGENE AUTOLEUCEL | 2022 | RRMM at least four prior lines including IMiD, PI, and aCD38 mAb |
IMiD, immunomodulatory drugs; IV, intravenous; PI, proteosome inhibitor; SC, subcutaneous; RRMM, relapsed and refractory multiple myeloma; NDMM, newly diagnosed multiple myeloma; R, revlimid (lenalidomide); d, dexamethasone; V, velcade (bortezomib); P, pomalidomide; T, thalidomide; K, kyprolis (carfilzomib); aCD38 mAB, anti-CD38 monoclonal antibody.
FDA-approved next-generation therapeutics
Monoclonal antibodies
CD38 -directed mAbs: daratumumab (Darzalex® and Darzalex Faspro®) and isatuximab (Sarclisa®)
Daratumumab (human) and isatuximab (chimeric) are IgG1κ mAb targeting CD38.3,4 Single-agent intravenous (IV) daratumumab was FDA-approved in November 2015 and subcutaneous (SC) and IV daratumumab are approved in triplet and quad-regimens for the treatment of patients with newly diagnosed (NDMM) and relapsed and refractory (RRMM) multiple myeloma. Isatuximab was approved for the treatment of RRMM in combination with pomalidomide and dexamethasone, and carfilzomib and dexamethasone in 2020 and 2021, respectively (Table 1). While both are available in IV formulations, SC daratumumab and hyaluronidase human-fihj (Darzalex Faspro®) was approved in 2020 based on the COLUMBA trial that demonstrated the non-inferiority of the SC formulation.5–7 Isatuximab SC is currently being investigated (NCT04045795). 8 In practice, prescribers can use both forms of daratumumab interchangeably, but SC is generally preferred based on more convenient administration, including decreased time of drug administration, first-dose adverse reactions and post-first-dose monitoring requirements, and overall improved patient satisfaction.9–12 A selection of relevant clinical trials investigating daratumumab and isatuximab as single agents and in combination regimens highlights the most common any grade and grade ⩾ 3 adverse events associated with aCD38 mAb treatment (Table 2).13–22
Table 2.
Selected trials investigating monoclonal antibodies and most relevant toxicities.
Trial | Regimen | Median prior lines (range) | Patient population | Most common any grade toxicities (non-SOC arm) | > Grade 3 events |
---|---|---|---|---|---|
DARATUMUMAB | |||||
Monotherapy 19 | 5 (2–14) | 95% PI + IMiD refractory, 31% quad-refractory | IRR (42%), fatigue (40%), anemia (33%), nausea (29%), thrombocytopenia (25%), neutropenia (23%), back pain (22%), cough (21%) | Anemia (24%), thrombocytopenia (19%), neutropenia (12%), IRR (5% – G3 only) | |
D-RVd vs RVd 22 | 0 | NDMM | Fatigue (69%), URI (63%), neuropathy (60%), diarrhea (60%), neutropenia (58%), constipation (52%), cough (51%), nausea (50%), fever (46%), thrombocytopenia (43%), IRR (42%), anemia (35%) | Neutropenia (41%), thrombocytopenia (16%), anemia (9%), neuropathy (7%), diarrhea (7%), fatigue (6%), IRR (6% – G3 only) | |
DRd vs Rd 14 | 1 (1–11) | 20% PI-refractory 3.5% IMiD-refractory 2.4% Dual refractory |
Neutropenia (59%), IRR (47%), diarrhea (43%), fatigue (35%), URI (32%), anemia (31%), constipation (29%), cough (29%), thrombocytopenia (27%), muscle spasms (26%), nausea (24%), fever (20%) | Neutropenia (52%), thrombocytopenia (27%), anemia (12%), pneumonia (8%), fatigue (6%), diarrhea (5%), IRR (5% – G3 only) | |
DKd vs Kd 15 | 2 (1–2) | 32% Len-refractory 28% Velcade-refractory |
Thrombocytopenia (37%), anemia (33%), HTN (31%), diarrhea (31%), URI (29%), fatigue (24%), dyspnea (20%) | Thrombocytopenia (24%), HTN (18%), anemia (17%), pneumonia (12%), neutropenia (9%) | |
DPd vs Pd 18 | 2 (1–5) | 79% Len-refractory 47% PI-refractory 42% Dual refractory |
Neutropenia (68%), infections (65%), anemia (37%), thrombocytopenia (33%), fatigue (25%), diarrhea (22%), fever (20%), IRR (5%) | Neutropenia (68%), infections (24%), thrombocytopenia (18%), anemia (17%), fatigue (8%) | |
ISATUXIMAB | |||||
Monotherapy 20 | 5 (1–13) | 100% IMiD + PI exposed | Anemia (98%), leukopenia (77%), thrombocytopenia (64%), neutropenia (45%), AST increase (43%), fatigue (37%), nausea (32%), ALT increase (29%), cough (23%), URI (24%), diarrhea (20%), dyspnea (19%) | Lymphopenia (34%), anemia (20%), thrombocytopenia 17%), neutropenia (12%), pneumonia (7%) | |
IsaKd vs Kd 21 | 2 (1–2) | 32% Len-refractory 20% Dual refractory |
Anemia (99%), thrombocytopenia (94%), respiratory infection (83%), neutropenia (55%), IRR (46%), HTN (37%), diarrhea (36%), pneumonia (29%), fatigue (28%), dyspnea (28%), thromboembolic events (15%), cardiac failure (7%) | Respiratory infection (32%), thrombocytopenia (30%), pneumonia (21%), anemia (20%), HTN (20%), neutropenia (19%) | |
IsaPd vs Pd 13 | 3 (2–4) | 94% Len-refractory 77% PI-refractory 72% dual refractory |
Anemia (99%), neutropenia (96%), thrombocytopenia (84%), IRR (38%), URI (28%), diarrhea (26%), pneumonia (20%), fatigue (17%), constipation (16%), nausea (15%) | Neutropenia (85%), anemia (32%), thrombocytopenia (31%), pneumonia (16%) | |
ELOTUZUMAB | |||||
EloRd vs Rd 17 | 2 (1–4) | 68% bortezomib exposed 5% lenalidomide exposed |
Infections (84%), diarrhea (50%), fatigue (49%), anemia (44%), fever (41%), constipation (36%), cough (34%), muscle spasm (31%), edema (30%), pneumonia (22%) | Lymphopenia (79%), neutropenia (34%), infections (33%), anemia (20%), thrombocytopenia (19%), pneumonia (14%), fatigue (10%), diarrhea (8%) | |
EloPd vs Pd 16 | 3 (2–8) | 90% Len-refractory 78% PI-refractory 68% dual refractory |
Infections (65%), anemia (25%), neutropenia (23%),constipation (22%), diarrhea (18%), thrombocytopenia (15%), respiratory tract infection (17%) | Infections (13%), neutropenia (13%), anemia (10%), thrombocytopenia (7%), pneumonia (5%) |
ALT, alanine aminotransferase; AST, aspartate transaminase; IMiD, immunomodulatory drugs; IRR, Infusion-related reactions; PI, proteosome inhibitor; URI, upper respiratory tract infections; infections; HTN, hypertension; NDMM, newly diagnosed multiple myeloma, SOC, standard of care; Len, lenalidomide.
Daratumumab is infused intraveneously at a dose of 16 mg/kg and injected over 3–5 min at a fixed dose of 1800 mg weekly for two cycles, every other week for four cycles, and then monthly thereafter. In contrast, isatuximab is infused at a dose of 10 mg/kg weekly for cycle 1, followed by every other week thereafter. The infusion time for daratumumab is based on a progressive titration starting with 6.5 h at first dose followed by 4.5 h at second dose and 3.5 h thereafter, whereas isatuximab is slowly titrated up for the first two weekly injections and typically lasts 3–4 h barring any reactions or complications. To decrease infusion times, numerous trials have confirmed that rapid infusion of daratumumab 90 min and isatuximab 70 min can be initiated safely following evidence of tolerability during the first cycle (Table 3).23–29
Table 3.
Practical considerations associated with monoclonal antibody treatment.
Practical considerations – monoclonal antibodies | ||||
---|---|---|---|---|
Daratumumab | Daratumumab Faspro | Isatuximab | Elotuzumab | |
Route | IV | SC | IV | IV |
Dosing | 16 mg/kg weekly C1–2, q2w C3–6 → q28 days | 1800 mg weekly C1–2, q2w C3–6 → q28 days | 10 mg/kg weekly for cycle 1 and then every other week thereafter | 10 mg/kg once weekly in cycles 1–2, then: ERd: 10 mg/kg every 2 weeks EPd: 20 mg/kg every 4 weeks |
Infusion/injection rates | First infusion 16 mg/kg (in 1 L) starting at 50 ml/h and increasing by 50 ml/h to a maximum of 200 ml/h Subsequent infusions 20% dose over 30 min (200 ml/h) → 80% over 60 min (450 ml/h) |
All injections 3–5 min SC injection |
First infusion 25 ml/h for 1 h then increase by 25 ml/h q30 min to maximum rate of 150 ml/h Second infusion 50 ml/h for 30 min then increase by 100 ml/h to maximum rate of 200 ml/h Subsequent infusions 200 ml/h |
10 mg/kg C1D1 0–30 min @ 0.5 ml/min, 30–60 min @ 1 ml/min, 60 min @ 2 ml/min 10 mg/kg C1D8 0–30 min @ 3 ml/min, ⩾ 30 min @ 4 ml/min 10 mg/kg C1D15 + 5 ml/min 20 mg/kg C1D1 0–30 min @ 3 ml/min, ⩾ 30 min @ 4 ml/min 20 mg/kg C1D8 + 5 ml/min |
Rapid infusion | C1D1 (7 h)–C1D15 (90 min) | NA | C1D1 (3.7 h)–C1D15 + (75 min) | C1D1 (2 h 50 min) to C1D15 + (53 min) |
IRR | ||||
Rates of IRR (%) (All grade/grades 3–4) |
Dd: 35/5 DRd: 47/5 |
Dd: 13/2 DPd: 5/0 |
IsaPd: 38/3 (98% resolved on C1D1) IsaKd: 46/1 (74% resolved on day of reaction) |
ERd: 11/1 EPd: 5/0 |
Time of onset | Dd: 1.5 h (range 1–24.5 h) DRd: 92% occurred with C1D1 |
Dd: 3.4 h (range 1–47.8 h) | IsaPd: During C1D1 infusion, no delayed
reactions IsaKd: Primarily during C1D1/C1D8 |
ERd: 70% occurred with dose 1 |
Prophylaxis 15–60 min prior to treatment | First infusion Acetaminophen, diphenhydramine, dexamethasone, montelukast, famotidine Subsequent infusions Famotidine, montelukast (discontinued after cycle 1) |
All infusions H2 blocker, diphenhydramine, acetaminophen, dexamethasone |
||
Dexamethasone dosing (If > 75 years, 50% reduction) |
First dose 20 mg IV C1D1 15–60 min prior to dose and 20 mg PO C1D2 Subsequent dosing 40 mg PO 1–3 h prior to dose |
C1D1 40 mg IV ~60 min prior to dose C1D8+ 40 mg PO 1–3 h prior to dose |
||
First-dose monitoring | 2 h after infusion if reactions occurs | 2 h maximum after C1D1 | 2 h after infusion if reaction occurs | |
Neutropenia | G-CSF considered in patients with grade ⩾ 3 neutropenia, especially early in treatment course for continued dosing. Reasonable to give 3 consecutive days of G-CSF with ANC < 500 cells/uL and 1 day with ANC 500–1000 cells/uL. Treatment should be withheld when a patient has a documented infection and re-initiation of treatment is based on severity of infection and resolution of symptoms | |||
Infection prophylaxis | Check hepatitis B, screen for HIV, and
hepatitis C as indicated Acyclovir 400 mg bid or valacyclovir 500 mg bid Levofloxacin + TMP-SMX DS if ALC < LLN + /– heavily pretreated |
|||
Hypogammaglobulinemia | IVIG if IgG < 400 and recurrent infections | |||
COVID | Recommend three (Johnson & Johnson: one dose for primary series + two boosters) or four doses (Pfizer-BioNTech and Moderna: two doses for primary series + two boosters) of vaccine and Evusheld for all actively treated patients |
IV, intravenous; IVIG, intravenous immunoglobulins; EPd, Elo + pomalidomide; ERd, Elo + lenalidomide; IRR, infusion-related reactions; SC, subcutaneous; ALC, absolute lymphocyte count; ANC, absolute neutrophil count; Dd, daratumumab + dexamethasone; DRd, daratumumab + lenalidomide + dexamethasone; DPd, daratumumab + pomalidomide + dexamethasone; IsaPd, isatuximab + pomalidomide + dexamethasone; IsaKd, isatuximab + carfilzomib + dexamethasone; G-CSF, granulocyte colony stimulating factor; DS, double strength; LLN, lower limit of normal; IgG, Immunoglobulin G; PO, by mouth; TMP-SMX, trimethoprimsulfamethoxazole.
Infusion-related reactions (IRRs) are common in all formulations but more likely with IV administration and occur primarily with the first dose (Table 3). Associated symptoms are generally low grade and include chills, fever, nausea, nasal congestion, cough, and dyspnea. The time to IRR onset is generally longer with SC versus IV daratumumab (3.4 versus 1.5 h, respectively), though low-grade, non-serious delayed IRR (incidence of IRR occurring the day after infusion/injection) have been reported for both daratumumab formulations but are rare.5,6 In general, those patients without IRR following IV daratumumab or isatuximab will not require post-dose monitoring, but if reactions are evident, we generally monitor patients for an additional 2 h after the completion of infusion. Patients treated with SC daratumumab are currently monitored for up to 4 h after the first dose and 1–2 h after the second dose though a recent retrospective study suggests that standard 30-min monitoring post-injection may be sufficient if adequate IRR prophylaxis is provided. 10
To mitigate the risk of IRR, split dosing may be considered with use of IV daratumumab, 30 however, utilization of an appropriate IRR prophylactic regimen prior to treatment with IV/SC daratumumab and isatuximab is essential to ensure safe drug administration. We recommend acetaminophen, diphenhydramine, dexamethasone, montelukast, and famotidine 15–60 min prior to drug administration (Table 3).31–33 After the initial cycle of either daratumumab formulation, famotidine and montelukast may be discontinued and a rapid taper of dexamethasone can be considered, especially in those patients that did not experience IRR, are steroid intolerant, or are receiving SC formulation.24,34,35 In addition, we do not recommend > 20 mg weekly dexamethasone or equivalent at any time for patients > 75 years of age. If IV daratumumab is preferred per patient preference or is used to reduce the minimal risk of delayed IRR associated with SC daratumumab, oral corticosteroids the day following infusion may be considered during the first cycle. In those patients treated with isatuximab, we utilize a similar prophylactic approach, though montelukast is not included and use of an H2 blocker is continued over the course of treatment. 29
Other than IRR, patients often experience hematologic toxicities, fatigue, dyspnea, upper respiratory tract infections (URI), including pneumonia, and gastrointestinal events, including diarrhea and constipation; the incidence and severity are ultimately dependent on the combination regimen used (Table 2). 36 Used as single agents, the most common all-grade treatment emergent adverse events (TEAEs) were URIs and arthralgias the addition of PIs are associated with higher incidence of peripheral neuropathy and thrombocytopenia, while the addition of IMiDs lead to more diarrhea, anemia, and neutropenia.13,15,18,19,21,22,37–40 In those patients treated with single-agent daratumumab or isatuximab, cytopenias (neutropenia, lymphopenia, anemia, and thrombocytopenia) were the most common grade 3–4 TEAEs, occurring primarily within the initial months of treatment initiation, and as expected, treatment with multi-agent regimens increased the incidence of these high-grade hematologic events. Notably, a prespecified subgroup analysis of COLUMBA indicated a higher incidence of neutropenia in patients with low body mass index (⩽ 65 kg) treated with SC daratumumab, however, this did not lead to a clinically notable increase in infections.5,41
Given the high incidence of associated neutropenia, lymphopenia, and hypogammaglobulinemia, patients are highly susceptible to bacterial and viral infections, most notably URI and pneumonia (Table 2). 36 Though data are limited, median time to first infection in a small case series of daratumumab treated patients was 2.5 months; range, 0.1–18.7 months. 42 Atypical infections are uncommon, however, reactivation of herpes zoster and Epstein–Barr virus/cytomegalovirus, pneumocystis jirovecii pneumonia (PJP), progressive multifocal leukoencephalopathy, bronchopulmonary aspergillosis, fungal meningitis, listeriosis, and disseminated cryptococcosis are reported.43–50 As expected, infections occur at a higher rate in those patients with higher grade neutropenia and lymphopenia, notably heavily pretreated patients treated with combination regimens within the initial 6 months of treatment. In this population treated with daratumumab, the median time to severe infection was ~50 days in patients with severe lymphopenia versus ~90 days in those without severe lymphopenia. 51
Given the increased risk of infection in these patients, all patients should receive antiviral prophylaxis with acyclovir or valacyclovir, PJP prophylaxis in the setting of prior PJP infection, heavily pretreated disease, or if the absolute lymphocyte count is below the lower limit of normal, and should be considered for intravenous immunoglobulins (IVIGs) in the setting of recurrent infections and IgG < 400 as this intervention has been associated with a 72% reduction in grade 3–4 infections. 42
Another important infectious consideration is the potential for hepatitis B virus (HBV) reactivation related to drug-associated natural killer (NK) cell depletion and suppression of humoral immunity.52–54 We recommend all patients be tested for HBV prior to starting mAb treatment and to undergo serial monitoring of liver enzymes over the course of treatment. Patients may continue mAb treatment alongside prophylaxis or preemptive therapy with either tenofovir or entecavir (not lamivudine due to low-resistance thresholds with this agent). 55 Monitoring and treatment of active hepatitis B disease requires consultation with a hepatologist to determine duration of therapy, appropriate frequency for trending viral load with PCR throughout treatment, and plan for maintaining prophylaxis against HBV once mAb therapy is completed. Patients with significant risk factors should also be screened for hepatitis C and HIV as clinically indicated. 56
Patients with MM are inherently at greater risk for infection and appropriate prevention strategies during the COVID-19 pandemic continue to be center stage for patients, family members, and care teams given this populations increased risk of adverse outcomes, including death.57–59 There is little doubt that the best protection from COVID-19 infection is prevention. Mask-wearing, hand-washing, avoidance of crowds, and sick individuals, and vaccination, even in patients potentially unable to mount an optimal vaccine response, are essential precautionary measures in all patients. We strictly follow the Centers for Disease Control guidelines for vaccination and encourage all of our patients to receive a full vaccination series, including the second booster. In addition, all patients are offered tixagevimab and cilgavimab (Evusheld™), a recombinant human IgG1κ mAb product that binds to non-overlapping epitopes of the spike protein receptor-binding domain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and blocks attachment to the human ACE2 receptor, for pre-exposure prophylaxis of COVID-19. 60
SLAMF7-directed antibody: elotuzumab (Empliciti®)
Elotuzumab targets signaling lymphocytic activation molecule F7 (SLAMF7) or CS-1, a glycoprotein expressed on the surface of more than 95% of bone marrow myeloma and NK cells. 61 Elotuzumab is an IV infusion first approved in 2015 in combination with lenalidomide and dexamethasone based on the ELOQUENT-2 trial for patients previously treated with one to three prior lines of therapy,17,38,39 and in 2018, was approved in combination with pomalidomide and dexamethasone (EPd) based on the ELOQUENT-3 trial.16,62 Treatment with both triplet regimens is based on 28-day cycles and elotuzumab is infused weekly for two cycles. Starting in cycle 3, when combined with lenalidomide, elotuzumab is infused every other week but in combination with pomalidomide, it is only infused on the first day of each cycle (Table 2).
Elotuzumab is associated with a much lower rate of IRR than the IV formulations of daratumumab and isatuximab. Data from the ELOQUENT-2 and 3 trials demonstrated a 5–11% rate of all-grade IRR, events were all grade 1 or 2 except for five patients that had a grade 3 event on ELOQUENT-2.16,39 Similar to daratumumab and isatuximab, the majority of these events were associated with fever, chills, cough, congestion, and nausea. 38 Similar to the other mAb, pretreatment with acetaminophen, diphenhydramine, famotidine, and dexamethasone is indicated 15–60 min prior to infusion, though different from the recommended dosing per label, we utilize standard dexamethasone dosing of either 20 or 40 mg based on age. 32 Other TEAE related to elotuzumab are similar to the anti-CD38 mAb and vary depending on the combination treatment regimen. For example, the most common grade 3 or higher events of triplet therapy included lymphopenia (77%), neutropenia (34%), infections (33%), anemia (20%), pneumonia (14%), fatigue (10%), and diarrhea (8%) on ELOQUENT-2, and infections (13%), neutropenia (13%), anemia (10%), thrombocytopenia (7%), and pneumonia (5%) on ELOQUENT-3 (Table 2). Our approach to IRR and infection prophylaxis associated with elotuzumab is similar to that described for the anti-CD38 mAb (Table 3).
B-cell maturation antigen-directed novel immunotherapeutics
ADC: belantamab mafodotin (BLENREP®)
Belantamab mafodotin is a first-in-class immunoconjugate or ADC consisting of an afucosylated humanized IgG1 B-cell maturation antigen (BCMA)-directed mAb conjugated to the microtubule-disrupting agent monomethyl auristatin F (MMAF). Belantamab mafodotin was FDA-approved in 2020 for patients with RRMM previously treated with ⩾ 4 prior therapies, including an IMiD, PI, and an anti-CD38 mAb based on the DREAMM2 trial, a randomized phase II trial investigating two different doses of belantamab in heavily pretreated patients median of seven prior lines of treatment.63,64 Based on the efficacy and safety profile, single-agent belantamab 2.5 mg/kg infused every 3 weeks was approved. Overall, it is considered a generally well-tolerated, steroid-sparing treatment, though it is associated with manageable corneal and hematologic toxicities.
DREAMM2 confirmed that microcyst-like epithelial changes (MECs) were common. 65 In those patients treated at the 2.5 mg/kg dose on DREAMM2, 72% of patients had any grade keratopathy (46% grade ⩾ 3) with or without symptoms or changes in best-corrected visual acuity (BCVA) that occurred on average 37 days (range, 19–143 days) after starting treatment. In addition, 54% had any grade (31%, grade ⩾ 3) BCVA changes, 18% had a meaningful decline in the Snellen Visual Acuity to 20/50 or worse that recovered within an average of 21.5 days, 25% had any grade blurred vision (4%, grade ⩾ 3), and 15% experienced any grade dry eyes (1%, grade ⩾ 3).63,65 In those patients with grade ⩾ 2 keratopathy, 77% recovered on average 86.5 days; range, 8–365 days after their initial ocular event. 65 No patient had permanent vision changes or loss, likely accounted for by the continuous regeneration of the corneal epithelial layer.64,65 Notably, in responding patients, 80% maintained their response even after dose holds over 63 days, 66 indicating that responses are durable even when drug dosing is modified.
Ultimately, clinical investigation of alternate dosing regimens will be instrumental in defining the best tolerated and efficacious treatment schedule, 67 but at this time, per requirements of the BLENREP REMS Program, treatment requires corneal examinations by an ophthalmologist or optometrist at baseline and before each treatment that include, at a minimum, assessment of BCVA and keratopathy by slit lamp examination. Use of preservative free eye drops (available for free through the GlaxoSmithKline BLENREP Eye Drop Supportive Care Program and avoidance of contact lenses while on treatment are also important practical considerations (Table 4). 68
Table 4.
Belantamab mafodotin practical considerations.
Practical considerations – belantamab mafodotin | |
---|---|
![]() |
Patients are required to have ophthalmic examinations (visual acuity and slit lamp) at baseline within 3 weeks prior to first dose, within 2 weeks prior to each dose, and for acute changes based on the FDA-mandated Risk Evaluation and Mitigation Strategy (REMS) Program |
![]() |
Preservative-free lubricant eye drops should be used 4 × daily (Eye Drop Program) |
![]() |
Patients should avoid wearing contact lenses while on treatment |
![]() |
Communication between patient and care team is key! |
![]() |
Patients should be advised prior to treatment that they may not be able to drive and in more severe cases, may not be able to read |
Other noteworthy belantamab-associated all-grade toxicities include cytopenias (thrombocytopenia (38%), anemia (27%), and lymphopenia (14%)), pyrexia (23%), fatigue (16%), and aspartate transaminase (AST) increase (21%), and the most common grade ⩾ 3 events include keratopathy, 46%; thrombocytopenia, 22%; anemia, 21%; lymphopenia, 13%; and neutropenia, 11%. 63 Overall, the toxicities associated with belantamab treatment are manageable, though ocular toxicities can be profound and special attention to management of ocular adverse events is critical for safe drug administration and further development of this agent in multi-drug combinations (Table 5).
Table 5.
Belantamab dose modifications.
Dose modifications for ocular toxicities | ||
---|---|---|
Severity | Corneal AE | Recommended action |
Grade 1 | Corneal exam: Mild superficial keratopathy BCVA: Decline from baseline of one line on Snellen Visual Acuity |
Continue treatment |
Grade 2 | Corneal exam: Moderate superficial
keratopathy BCVA: Decline from baseline of two or three lines on Snellen and not worse than 20/200 |
Hold treatment until improvement to < G1 and resume same dose |
Grade 3 | Corneal exam: Severe superficial
keratopathy BCVA: Decline from baseline by > 3 lines on Snellen and not worse than 20/200 |
Hold treatment until < G1 and resume at reduced dose |
Grade 4 | Corneal exam: Corneal epithelial defect BCVA: Snellen Visual Acuity worse than 20/200 |
Consider permanent discontinuation. If continuing, hold treatment until < G1 and resume at reduced dose |
BCVA, best-corrected visual acuity; AE, adverse event
CAR-T: chimeric antigen receptor T-cells; idecabtagene vicleucel (Abecma®, Ide-cel) and ciltacaptagene autoleucel (Carvykti®, Cilta-cel)
The clinical development of multiple b-cell maturation antigen (BMCA)-directed CAR-T constructs, namely, Ide-cel and Cilta-cel, highlight two of the many novel T-cell adoptive therapies available commercially and in clinical trial. Ide-cel was FDA-approved in 2021, based on the KARMMA platform, for patients previously treated with at least four prior lines of therapy, including an IMiD, PI, and anti-CD38 mAb. 69 Cilta-cel, developed on the CARTITUDE platform, is associated with impressive responses, 70 and was recently FDA-approved in early 2022 (Table 6). The toxicities associated with CAR-T in RRMM reflect the CAR-T experience in other hematologic malignancies and are characterized by cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). In addition, cytopenias, including B-cell aplasia, hypogammaglobulinemia associated with the need for infection prophylaxis, and hemophagocytic lymphohistiocytosis (HLH) are major considerations in routine practice (Table 7).
Table 6.
Selected next generation clinical trials investigating novel immunotherapeutics.
Agent | Trial | Target epitope | N | All grade/> G3 (%) | mTTO (days) |
All grade/> G3 (%) | mTTO (days) |
Toci | Toxicities of interest: All grade | Toxicities of interest: > Grade 3 events |
---|---|---|---|---|---|---|---|---|---|---|
CAR-T | ||||||||||
Ide-cel | KARMMA | BCMA | 128 | 84/5 | 1 | 18/3 | 2 | 52% | Neutropenia (91%), CRS (84%), anemia (70%), thrombocytopenia (63%), leukopenia (42%), diarrhea (35%), fatigue (34%), decreased appetite (21%), headache (21%) | Neutropenia (89%), anemia (60%), thrombocytopenia (52%), leukopenia (39%), hypophosphatemia (16%) |
Cilta-cel | CARTITUDE | 97 | 95/4 | 7 | 21/9 | 8 | 69% | Neutropenia (96%), CRS (95%), anemia (81%), thrombocytopenia (79%), leukopenia (62%), fatigue (37%), cough (35%), diarrhea (30%), decreased appetite (29%) | Neutropenia (95%), anemia (68%), thrombocytopenia (60%), leukopenia (61%), NT (9%), hypophosphatemia (7%) | |
T-CELL REDIRECTING BISPECIFIC ANTIBODIES | ||||||||||
AMG701 | ParadigMM | BCMA | 82 | 65/9 | NR | NR | NR | 29% | CRS (65%), anemia (42%), diarrhea (31%), hypophosphatemia (31%), neutropenia (25%), thrombocytopenia (21%) | NR |
Teclistamab | MajesTEC-1 | 40* | 70/0 | 1 | 1 patient – G1 | NR | 35% | CRS (70%), neutropenia (65%), anemia (50%), thrombocytopenia (45%), fatigue (38%), nausea (33%), diarrhea (23%), headache (20%) | Neutropenia (40%), anemia (28%), thrombocytopenia (20%), leukopenia (18%) | |
Elranatamab | MagnetisMM-1 | 30 | 73/0 | 1 | 20/0 | NR | 30% | Lymphopenia (83%), CRS (73%), anemia (60%), neutropenia (53%), thrombocytopenia (53%), injection site reaction (50%), nausea (37%), increased AST/ALT (33/30%), diarrhea (30%) | Lymphopenia (83%), neutropenia (53%), anemia (50%), thrombocytopenia (37%), increased AST/ALT (10%), nausea (3%) | |
ABBV-383B | NCT03933735 | 58 | 45/0 | < 1 | NR | NR | 9% | CRS (45%), fatigue (24%), headache (22%), anemia (21%), infection (21%), nausea (21%), neutropenia (19%), thrombocytopenia (17%), fever (16%) | Anemia (17%), neutropenia (16%), thrombocytopenia (14%), infection (14%) | |
REGN5458 | NCT03761108 | 49 | 39/0 | < 1 | 12/0 | NR | 32% | Infections (47%), CRS (39%), anemia (37%), fatigue (35%), nausea (31%), pyrexia (31%), back pain (27%), thrombocytopenia (18%), neutropenia (16%) | Anemia (22%), infections (18%), neutropenia (14%), lymphopenia (12%), fatigue (6%), thrombocytopenia (6%) | |
Talquetamab | MonumenTAL-1 | GPRC5D | 30* | 73/2 | 2 | 7/0 | NR | 60% | CRS (73%), neutropenia (67%), dysgeusia (60%), anemia (57%), leukopenia (37%),dysphagia (37%), thrombocytopenia (33%), fatigue (30%), nausea (23%) | Neutropenia (60%), lymphopenia (30%), anemia (27%), leukopenia (27%), thrombocytopenia (20%), CRS (2%), pyrexia (2%) |
Denotes number of patients treated at the recommended phase 2 dose (RP2D) and associated toxicity profile. ALT, alanine aminotransferase; AST, aspartate transaminase; BCMA, B-cell maturation antigen; CAR-T, chimeric antigen receptor T; CRS, cytokine release syndrome; mTTO, median time to onset; NR, not reported; NT, neurotoxicity.
Table 7.
Practical considerations associated with CAR-T and bispecific T-cell engager.
Practical considerations – CAR-T and T-cell redirecting bispecific antibodies | ||
---|---|---|
AE | CAR-T | T-cell redirecting bispecific antibody |
CRS | • Grade 1 – Supportive care with
consideration of tocilizumab for early onset fevers
(< 72 h) • Grade 2 – Tocilizumab 8 mg/kg IV and increase vital sign monitoring from every 4 to every 2 h • Grade 3 or 4 – Tocilizumab 8 mg/kg IV and transfer to ICU • If improvement after initial treatment, continue supportive care and if on steroids, stop after 3 days or taper as appropriate • If no improvement/worsening, repeat tocilizumab for up to 4 doses, add dexamethasone 10 mg IV q6 h and rule out alternative etiologies • If symptoms do not improve after —three to four doses of tocilizumab, increase steroid to methylprednisolone 1000 mg IV daily and add anakinra SQ/IV 100–400 mg/day × 7 days • If no improvement evident after 7 days, consider siltuximab 11 mg/kg IV over 1 h • If no improvement, consider T-cell ablation with cyclophosphamide 1.5 g/m2 IV |
|
ICANS/NT | • Grade 1 – Start levetiracetam 500 mg PO
bid and continue until day 30 and ICANS
grade 0, neurologic exams every 8 h, rule out
alternative etiologies, and consider starting
dexamethasone 10 mg once • Grade 2 – Treat as grade 1 with the addition of dexamethasone 10 mg IV every 6 h, neurologic exams every 4 h, and consider work-up with (1) MRI brain with and without contrast, (2) lumbar puncture with opening pressure, cultures, protein, IL-6, oligoclonal bands, and cell count, and (3) 30-min EEG, and treat seizures as needed • Grade 3 or 4 – Confirm grade 1 and 2 orders are completed, ICU admission, neurologic exams every hour, and consider (1) continuous EEG and neurology consultation, (2) airway protection, and (3) discontinuation of anticoagulation |
|
Cytopenias | G-CSF/transfusions/TPO-RA | |
Marrow failure | Consider stem cell boost if alternative etiologies (TTP/HUS/hemolysis) are ruled out and repeat bone marrow biopsy is significantly hypocellular/acellular > day + 60 | NA |
Hypogammaglobulinemia | IVIG if IgG < 400–600 and recurrent infections | |
Antibiotic prophylaxis | Acyclovir, levofloxacin, and fluconazole (ANC < 500), TMP-SMX (CD4 < 200)a | NA |
Mold fungal prophylaxisb | Discontinue fluconazole, add posaconazole or voriconazole | NA |
COVID-19 | SOC vaccination starting at day + 90 and Evusheld ~day + 15–30 if no CRS/ICANS | SOC vaccinations and Evusheld |
aIf CD4 T-cell count < 200 uL, continue until CD4 is consistently > 200 uL. bIf > 1 dose of tocilizumab, > 3 days steroids (> 10 mg dexamethasone or equivalent), and/or use of 2nd line agents for CRS (eg. anakinra, siltuximab). CAR- T , c himeric antigen receptor T; CRS, cytokine release syndrome; EEG, electroencephalogram; ICANS, immune effector cell-associated neurotoxicity syndrome; ICU, intensive care unit; IV, intravenous; IVIG, intravenous immunoglobulins; MRI, magnetic resonance imaging; ANC, absolute neutrophil count; G-CSF, granulocyte colony stimulating factor; HUS, hemolytic uremic syndrome; IgG, immunoglobulin G, IL-6, interleukin-6, NT, neurotoxicity; PO, by mouth; SOC, standard of care; SQ, subcutaneous; TMP-SMX, trimethoprim-sulfamethoxazole; TPO-RA, thrombopoietin receptor agonist; TTP, thrombotic thrombocytopenia purpura.
CRS is a clinical syndrome resulting from induction of inflammatory cytokines associated with T-cell activation and proliferation. It is most commonly associated with relatively low-grade clinical manifestations, including fever, rigors, tachycardia, shortness of breath requiring oxygen, and hypotension. While the majority of patients experience low-grade toxicities per the Lee et al. 71 criteria, life-threatening symptoms with multi-organ involvement can occur. Based on the KARMMA experience, 84% of patients developed any grade CRS, 95% were grade 2 or less, and 52% received tocilizumab, a potent mAb inhibitor of the IL-6 receptor. The median time to CRS onset was 1 day; range, 1–12 days and symptoms lasted a median of 5 days; range, 1–63 days. 69 These findings are consistent with the CARTITUDE experience, in that 95% of patients treated with Cilta-cel experienced any grade CRS associated with 96% that had grade 2 or less events. Interestingly, the median time to CRS onset in these patients was 7 days (range, 5–8 days), median duration of CRS was 4 days (range, 3–6 days), and almost 70% of patients were treated with tocilizumab (Table 3). While tocilizumab is the most commonly used intervention for CRS, corticosteroids (usually dexamethasone) and anakinra, an IL-1 receptor antagonist, may also be utilized given their ability to suppress the inflammatory cascade.
HLH, also known as macrophage-activation syndrome (MAS), is another complication of CAR-T therapy and was present in 4–8% of patients in the Ide-cel dataset. 69 This entity is difficult to distinguish from CRS as both present with high fevers, organ toxicity, cytopenias, hyperferritinemia, and high levels of inflammatory cytokines. Often, onset is just after development of or in conjunction with CRS. Diagnosis is made with criteria described by the CARTOX working group, which is based on high ferritin levels > 10,000 and lack of response to treatment instituted for CRS. 72 In cases not responding to tocilizumab after 48 h, consideration is given for treatment of presumed HLH with anakinra or etoposide while further work-up is completed.
ICANS is the second most common immune cell-mediated toxicity associated with CAR-T. Clinical manifestations may include headache, aphasia, tremors or seizures, encephalopathy, and death resulting from cerebral edema. The pathology underlying ICANS/neurotoxicity is not well understood but may be related to cytokine crossover to the cerebrospinal space through disruption of the blood–brain barrier, allowing for immune cell trafficking into the central nervous system and further inflammatory cascade activation.73,74 In KARMMA, 18% of patients were reported to have all-grade ICANS/neurotoxicity of which 97% were ⩽ grade 2 and occurred at a median of 2 days (range, 1–10). 69 In CARTITUDE-1, 17% of patients were reported to have any grade ICANS with two patients experiencing a grade 3 or higher event. This occurred at a median of 8 days; range, 6–8 days. Perhaps uniquely, five patients reportedly developed movement disorders, neurocognitive events, and personality changes MNT after recovery from CRS and/or typical ICANS. 75 Common features of this group included high tumor burden, grade ⩾ 2 CRS, ICANS, and high CAR-T cell expansion/persistence. Lab features included higher absolute lymphocyte counts, absolute CD4+ T-cells and CAR-T cell persistence at days 14, 21, and 28. Higher peak levels of IL-6 and interferon gamma (IFN-γ) were also seen in this group. Treatment for ICANS includes first-line therapy with corticosteroids and then tocilizumab, siltuximab, or anakinra. Those receiving Cilta-cel and who develop MNT-associated symptoms may be less responsive to steroids, however, hence early stratification of these patients to receive other cytokine therapy beyond steroids/tocilizumab, appropriate full neurologic and infectious evaluation, including CSF exam and seizure prophylaxis are recommended. 75
The risk of prolonged cytopenias, including B-cell aplasia, is not uncommon. Grade 3–4 cytopenias lasting greater than 30 days after CAR-T infusion has been reported in 20–40% of patients in patients with non-Hodgkin lymphoma (NHL) and acute lymphoblastic leukemia (ALL).76–80 In regard to Ide-cel, prolonged neutropenia was seen in 34–49% of patients depending on T-cell dose with 85% of patients taking a median of 1.9 months to recover counts. Prolonged thrombocytopenia occurred in almost half of the patients with a median duration of 2.1 months. There is currently no consensus on the routine use of growth factors and thrombopoietin receptor agonists, but both may be considered, especially in those with infections or bleeding tendencies. Consideration should be given to the separation of G-CSF from CAR-T products by at least 3 weeks or until resolution of CRS. If these interventions fail, CD34 + stem cell boosts may be a viable option for rescue after bone marrow and peripheral blood evaluation has ruled out HLH and other causes of cytopenias including thrombotic thrombocytopenic purpura/hemolytic uricemia syndrome.
Because of off-target effects resulting from B-cell aplasia, immunoglobulin levels may be suppressed in patients following CAR-T therapy and for this reason, patients remain at high risk for infections and associated complications. We recommend that family and caregivers receive influenza and COVID-19 vaccinations prior to infusion of lymphodepleting chemotherapy for this reason. In addition, antiviral and PJP prophylaxis are provided through 6 months or longer post-CAR-T infusion if the CD4 count remains < 200 cells/uL (checked monthly). The use of antibacterial and antifungals may be considered in cases of prolonged neutropenia. In those treated for CRS or ICANS with corticosteroids, tocilizumab, or other agents, we routinely add fungal prophylaxis for 30 days after the last dose. Immunoglobulin replacement therapy is also instituted routinely for our patients if their IgG level is below 400–600 mg/dl and checked on a monthly basis, especially if recurrent infections are present (Table 7).
Selective inhibitor of nuclear export (SINE): selinexor (Xpovio®)
Selinexor is an oral selective inhibitor of nuclear export blocking exportin 1 (XPO1), a nuclear exporter overexpressed in MM cells that shuttles tumor suppressor proteins, the glucocorticoid receptor, and oncoprotein messenger RNAs out of the nucleus.81,82 Selinexor was initially FDA-approved in 2019 for the treatment of patients previously treated with last least four prior lines of therapy, including lenalidomide, pomalidomide, bortezomib, carfilzomib, and an anti-CD38 mAb based on the STORM trial. 83 In 2020, based on the randomized phase III BOSTON trial, selinexor combined with bortezomib was approved for patients previously treated with at least one prior line of therapy 84 and a recent meta-analysis showed that selinexor combined with a PI not only improves response but also reduces the incidence of common all-grade toxicities. 85 Based on results from STOMP,86–89 the National Comprehensive Cancer Network (NCCN) guidelines now lists multiple selinexor-based combinations as ‘Useful in Certain Circumstances for Early Relapses (1–3 prior therapies)’, 56 supporting use of these regimens in routine clinical practice (Table 8).
Table 8.
Other selected next generation therapeutics.
Trial | Regimen | Median prior lines (range) | Most common any grade toxicities (non-SOC arm) | > Grade 3 events | Dose delay/reduction | Treatment discontinuation |
---|---|---|---|---|---|---|
SELINEXOR | ||||||
STORM | Monotherapy | 7 (3–18) | Thrombocytopenia (73%), fatigue (73%), nausea (72%), anemia (67%), decreased appetite (56%), decreased weight (50%), diarrhea (46%), neutropenia (40%), vomiting (38%), hyponatremia (37%), leukopenia (33%), URI (23%) | Thrombocytopenia (38%), anemia (43%), fatigue (25%), hyponatremia (22%), neutropenia (21%), leukopenia (14%), nausea (10%), pneumonia (9%) | 80% | 18% |
BOSTON | SVd vs Vd | 2 (1–2) | Thrombocytopenia (60%), nausea (50%), fatigue (42%), anemia (36%), diarrhea (32%), decreased appetite (35%), peripheral neuropathy (32%), weight loss (26%), asthenia (25%), vomiting (21%) | Thrombocytopenia (39%), anemia (16%), fatigue (13%), pneumonia (12%), neutropenia (9%), nausea (8%), asthenia (8%) | 89% | 21% |
STOMP | SKd | 4 (1–8) | Thrombocytopenia (72%), nausea (72%), anemia (53%), fatigue (53%), decreased appetite (47%), weight decrease (41%), leukopenia (34%), dysgeusia (31%), neutropenia (28%), diarrhea (25%), neuropathy (19%) | Thrombocytopenia (47%), anemia (19%), leukopenia (9%), fatigue (9%), neutropenia (6%), neuropathy (3%) | NR | 16% |
VENETOCLAX | ||||||
BELLINI | VenVd vs Vd | 2 (1–3) | Diarrhea (58%), nausea (36%), constipation (34%), fatigue (28%), peripheral neuropathy (29%), URI (29%), insomnia (28%), thrombocytopenia (26%), anemia (26%), neutropenia (23%), pneumonia (21%), cough (21%), emesis (19%) | Neutropenia (18%), anemia (15%), thrombocytopenia (15%), diarrhea (15%), pneumonia (14%), thrombocytopenia (11%) | 30% | 22% |
M13-367 | Monotherapy in t(11;14) | 5 (2–12) | Diarrhea (36%), lymphopenia (32%), nausea (26%), anemia (23%), cough (17%), neutropenia (16%), fatigue (16%), thrombocytopenia (10%), URI (10%) | Lymphopenia (19%), anemia (16%), thrombocytopenia (10%), sepsis (10%), neutropenia (7%), TLS (3%) | NR | NR |
M15-538 | VenKd | 1 (1–2) | Diarrhea (65%), fatigue (47%), nausea (47%), URI (39%), dyspnea (35%), cough (33%), emesis (29%), HTN (27%), pneumonia (18%) | Hypertension (16%), pneumonia (12%), insomnia, (10%), diarrhea (10%), influenza (6%) | NR | NR |
URI, upper respiratory tract infection; infection; HTN, hypertension; NR, not reported; SOC, standard of care; TLS, tumor lysis syndrome.
Selinexor combination therapy is associated with gastrointestinal toxicities, including nausea, vomiting, and diarrhea, decreased appetite and weight loss, fatigue, cytopenias, and URI. 85 As expected, the incidence and severity of these events vary based on selinexor dosing and combination therapy, and in general, patient education and significant supportive care, especially over the initial two cycles are required to maximize tolerability and drug exposure. 90
Appropriate dosing is critical and use of selinexor has now shifted from twice weekly to once weekly dosing when used in triplet regimens. 90 When used in combination triplet regimens, we recommend initiating selinexor 60–80 mg weekly during the first cycle and escalating to 100 mg weekly if tolerated. Supportive measures include use of low-dose olanzapine (2.5–5.0 mg) nightly for 3 days starting the day of selinexor, use of 5-HT3 receptor antagonist (most commonly ondansetron), and consideration of neurokinin-1 receptor antagonist (rolapitant). Based on drug tolerability, these anti-emetics may be weaned off following two cycles of treatment. We recommend obtaining at least weekly complete blood counts with dose holds and modifications as necessary, and G-CSF, transfusion support, and consideration of thrombopoietin receptor agonists and erythropoietin stimulating agents when applicable. Patients should also complete a once weekly metabolic panel. We instruct patients to maintain adequate hydration with at least 2 liters of fluid daily, ideally inclusive of salt-containing drinks, though in patients unable to tolerate this level of fluid intake, we consider use of salt tabs. In addition, we encourage patients to utilize anti-diarrheals (loperamide or diphenoxylate/atropine, or both) as necessary, consume adequate calories, consider use of appetite stimulants, and have a low threshold for consultation with a nutritionist. Ultimately, time and experience have proven that early and efficient supportive care leads to improved tolerability and allows patients to stay on treatment, if responding (Table 9).
Table 9.
Selinexor practical considerations.
Practical considerations – selinexor | |
---|---|
Condition | Supportive care |
Thrombocytopenia | Romiplostim 1–10 mcg/kg SC weekly |
Nausea/emesis | Ondansetron 8 mg bid – days 1–3 after each
dose* Olanzapine 2.5–5.0 mg PO qhs* 2 liters fluid intake daily (H2O + NaCl containing) +/– IVF +/– salt tabs Optional: NK1R antagonist PO • Rolapitant (reduces pill burden) • Aprepitant (increases concentration of dexamethasone) Cannabinoids |
Nutrition/anorexia | Olanzapine 2.5– 5.0 mg PO qhs* Consider boost or ensure Consult nutritionist |
Diarrhea | Anti-diarrheals prn |
Fatigue | Methylphenidate 5–10 mg daily |
Neutropenia | G-CSF (at appropriate timepoints for ANC < 1000) |
*Should be started in all patients before treatment and continued for 1-2 cycles; SC, subcutaneous; ANC, absolute neutrophil count; G-CSF, granulocyte colony stimulating factor; IVF, intravenous fluids; PO, by mouth; SOC, standard of care.
Next-generation non-FDA-approved therapeutics in clinical trials
T-cell redirecting bispecific antibodies
Current therapies aimed at overcoming the profound immune dysfunction associated with RRMM are proving highly effective even in heavily pretreated patients. T-cell redirecting bispecific antibodies are an off-the-shelf, steroid-sparing, novel immunotherapy drug class with great potential to change the MM treatment landscape. The constructs closest to routine clinical use are designed to simultaneously bind CD3 on T-cells and a specific target epitope on the MM cell the current majority target BCMA and are still investigational at this time, these include AMG701, 91 teclistamab, 92 elranatamab, 93 ABBV-383B, 94 and REGN5458, 95 while talquetamab targets GPRC5D 96 (Table 6). Despite the fact that there are currently no FDA-approved bispecific antibodies, these drugs are moving through phase I and II trials and are actively being investigated in randomized phase III trials.91,93,94,96–98
T-cell redirecting bispecific antibodies are associated with response rates in the 60–80% range even as single agents.91–98 As a drug class, they are generally well-tolerated and associated with a relatively consistent side effect profile, including significant all-grade cytopenias, nausea, diarrhea, fatigue, transient increases in aspartate transaminase (AST) and alanine aminotransferase (ALT) (Table 3), and risk of low-grade neurotoxicity. Low-grade CRS, however, is the hallmark of these drugs. All of these agents are associated with grade 1 and 2 events with a typical onset within 1–2 days of initiating treatment and generally recognized during ramp-up and with the initial full-strength dosing. The clinical presentation of these events is consistent with CAR-T therapy, however, intervention with tocilizumab with or without steroids is less frequently needed (Table 4). Current clinical investigations of the bispecifics aim to decrease CRS incidence using step-up dosing, shifting to SC injections rather than infusion, and utilization of early intervention with tocilizumab with or without steroids. If successful, these highly effective drugs will likely take over the early relapsed space and be investigated in newly diagnosed patients.
BCL-2 inhibitor: venetoclax (Venclexta®)
Venetoclax is an oral, small molecule, highly selective BH3 mimetic that induces apoptosis by displacing proapoptotic proteins from bcl-2 and is the first targeted therapy for the treatment of MM. 99 This bcl-2 inhibitor is not yet FDA-approved despite national guideline recommendations supporting its use in combination with dexamethasone for patients with t(11;14) disease. 56 Currently, clinical trials are investigating venetoclax in combination with bortezomib, carfilzomib, and daratumumab for patients with bcl-2-dependent disease harboring t(11;14) or high bcl-2 expression (Table 5).100–104
In general, patients treated with venetoclax commonly experience diarrhea, nausea, and cytopenias that are managed per standard of care measures. However, it is evident that single-agent and combination venetoclax-based regimens are associated with an increased risk of potentially life-threatening infections. The BELLINI trial was a randomized phase III trial that investigated venetoclax in combination with bortezomib and dexamethasone versus the bortezomib and dexamethasone. 104 Results from this trial indicated impressive responses, but the experimental arm was associated with an unexpectedly high mortality rate. Subanalysis of patients with t(11;14) disease showed improved responses, and the addition of venetoclax was not associated with increased rates of mortality in this population versus those treated with bortezomib and dexamethasone alone. Ultimately, the results of BELLINI halted the approval of venetoclax combination therapy though it substantiated its use in patients with t(11;14) disease. Based on these results and data derived from other trials with venetoclax-based combination regimens,101–103 high-grade pneumonia remains one of the most concerning TEAE. As such, the more recent M15-538 trial mandated antibiotic prophylaxis though rates of pneumonia did not differ from the venetoclax monotherapy experience. 93 Overall, appropriate patient selection and use of vigilant supportive care, most notably, early detection and treatment of pneumonia and gastrointestinal toxicities, may allow effective use of this novel therapy for patients with t(11;14) MM (Table 10).
Table 10.
Venetoclax practical considerations.
Practical considerations – venetoclax | |
---|---|
Consideration | Action |
Dosing | Full-dose (800 mg) venetoclax can be started on C1D1 without dose escalation as TLS is unlikely. 71 No dose adjustments are needed for renal insufficiency (not been studied in CrCl ⩽ 15). Dose reduction of 50% should be considered for severe hepatic impairment (Child-Pugh Class C) |
Infectious disease | Antibacterial (fluoroquinolone) and anti-PJP prophylaxis (TMP-SMX) may be considered, especially in the first two cycles, given increased incidence of pneumonia |
Diarrhea | Given the incidence of grade 3 or higher nausea and diarrhea, anti-diarrheals and anti-emetics should be strongly considered, especially in those patients that are frail or have a history of gastrointestinal toxicities with other treatment regimens |
Drug interactions | Notable drug-drug interactions include cardiac medications, such as carvedilol and amiodarone, and antifungals, including voriconazole and posaconazole |
PJP, pneumocystis jirovecii pneumonia; TLS, tumorlysis syndrome; TMP-SMX, trimethoprim sulfamethoxazole.
Conclusion
Since the FDA approval of daratumumab in 2015, the therapeutic options in the RRMM pipeline have greatly expanded. Incorporating these novel agents into established treatment combinations portends deeper responses and longer survival in the relapsed and refractory setting. Fortunately, many of these agents are less toxic than drugs historically used in the late-line setting and tolerable side effect profiles are allowing more patients access to a wider range of therapeutic options over the course of their disease. Driving into the future, utilization of our ever-expanding MM treatment toolbox and toxicity management will become increasingly complex as we continue to explore our armamentarium of novel therapeutics. While this massive momentum carries great promise for transforming myeloma into a chronic and treatable condition in many patients, consideration of interventions to offset the growing list of unique toxicities is paramount to enhancing both the quality and longevity of our patient’s lives. While this review provides a framework for the present and near future, there is no doubt that toxicity management strategies will continue to take center stage, and great attention must be paid to ensure that we maximize the quality of life of those patients under our care.
Acknowledgments
The authors express their special thanks to Claire Davies, MS, Strategic Communications at the Huntsman Cancer Institute, for her generosity and willingness to assist with graphic design. They also greatly appreciate the work of Michael Filtz, PharmD and Catherine Lee, MD, as they defined their institutional CRS/ICANS algorithms and graciously allowed them to include their work in this review.
Footnotes
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contributions: Mary Steinbach: Conceptualization; Data curation; Formal analysis; Writing – original draft; Writing – review & editing.
Kelley Julian: Conceptualization; Data curation; Formal analysis; Writing – original draft; Writing – review & editing.
Brian McClune: Conceptualization; Data curation; Formal analysis; Writing – original draft; Writing – review & editing.
Douglas W. Sborov: Conceptualization; Data curation; Formal analysis; Writing – original draft; Writing – review & editing.
ORCID iD: Mary Steinbach
https://orcid.org/0000-0002-9444-7166
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
Conflict of interest statement: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: M.S. is on the speakers’ bureau for GlaxoSmithKline, Karyopharm Therapeutics, and Janssen Pharmaceuticals. K.J. and B.L.M. have no relevant disclosures. D.W.S. is an advisor/consultant for Janssen Pharmaceuticals, Celgene, Abbvie, Sanofi, and GlaxoSmithKline.
Availability of data and materials: Not applicable.
Contributor Information
Mary Steinbach, Department of Internal Medicine, Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, USA.
Kelley Julian, Department of Pharmacy, The University of Utah, Salt Lake City, UT, USA.
Brian McClune, Department of Internal Medicine, Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, USA.
Douglas W. Sborov, Department of Internal Medicine, Huntsman Cancer Institute, The University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA.
References
- 1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2021. CA Cancer J Clin 2021; 71: 7–33. [DOI] [PubMed] [Google Scholar]
- 2. Joseph NS, Kaufman JL, Dhodapkar MV, et al. Long-term follow-up results of lenalidomide, bortezomib, and dexamethasone induction therapy and risk-adapted maintenance approach in newly diagnosed multiple myeloma. J Clin Oncol 2020; 38: 1928–1937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. De Weers M, Tai YT, Van Der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol 2011; 186: 1840–1848. [DOI] [PubMed] [Google Scholar]
- 4. Deckert J, Wetzel MC, Bartle LM, et al. SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clin Cancer Res 2014; 20: 4574–4583. [DOI] [PubMed] [Google Scholar]
- 5. Mateos MV, Nahi H, Legiec W, et al. Subcutaneous versus intravenous daratumumab in patients with relapsed or refractory multiple myeloma (COLUMBA): a multicentre, open-label, non-inferiority, randomised, phase 3 trial. Lancet Haematol 2020; 7: e370–e380. [DOI] [PubMed] [Google Scholar]
- 6. Usmani SZ, Nahi H, Legiec W, et al. Final analysis of the phase 3 non-inferiority COLUMBA study of subcutaneous versus intravenous daratumumab in patients with relapsed or refractory multiple myeloma. Haematologica. Epub ahead of print 31 March 2022. DOI: 10.3324/haematol.2021.279459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Usmani SZ, Nahi H, Mateos MV, et al. Subcutaneous delivery of daratumumab in relapsed or refractory multiple myeloma. Blood 2019; 134: 668–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Moreau P, Parmar G, Prince M, et al. A multi-center, phase 1b study to assess the safety, pharmacokinetics and efficacy of subcutaneous isatuximab plus pomalidomide and dexamethasone, in patients with relapsed/refractory multiple myeloma. Blood 2021; 138: 2744. [Google Scholar]
- 9. Hamadeh IS, Moore DC, Martin A, et al. Transition from intravenous to subcutaneous daratumumab formulation in clinical practice. Clin Lymphoma Myeloma Leuk 2021; 21: 470–475. [DOI] [PubMed] [Google Scholar]
- 10. Hughes DM, Henshaw L, Blevins F, et al. Standard 30-minute monitoring time and less intensive pre-medications is safe in patients treated with subcutaneous daratumumab for multiple myeloma and light chain amyloidosis. Clin Lymphoma Myeloma Leuk. Epub ahead of print 9 March 2022. DOI: 10.1016/j.clml.2022.03.003. [DOI] [PubMed] [Google Scholar]
- 11. Soefje S, Carpenter C, Carlson K, et al. Clinical administration characteristics of subcutaneous and intravenous administration of daratumumab in multiple myeloma patients at Mayo Clinic. Blood 2021; 138: 2717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Usmani SZ, Mateos MV, Hungria V, et al. Greater treatment satisfaction in patients receiving daratumumab subcutaneous vs. intravenous for relapsed or refractory multiple myeloma: COLUMBA clinical trial results. J Cancer Res Clin Oncol 2021; 147: 619–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Attal M, Richardson PG, Rajkumar SV, et al. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet 2019; 394: 2096–2107. [DOI] [PubMed] [Google Scholar]
- 14. Bahlis NJ, Dimopoulos MA, White DJ, et al. Daratumumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a randomized, open-label, phase 3 study. Leukemia 2020; 34: 1875–1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Dimopoulos M, Quach H, Mateos MV, et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study. Lancet 2020; 396: 186–197. [DOI] [PubMed] [Google Scholar]
- 16. Dimopoulos MA, Dytfeld D, Grosicki S, et al. Elotuzumab plus pomalidomide and dexamethasone for multiple myeloma. N Engl J Med 2018; 379: 1811–1822. [DOI] [PubMed] [Google Scholar]
- 17. Dimopoulos MA, Lonial S, Betts KA, et al. Elotuzumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended 4-year follow-up and analysis of relative progression-free survival from the randomized eloquent-2 trial. Cancer 2018; 124: 4032–4043. [DOI] [PubMed] [Google Scholar]
- 18. Dimopoulos MA, Terpos E, Boccadoro M, et al. Daratumumab plus pomalidomide and dexamethasone versus pomalidomide and dexamethasone alone in previously treated multiple myeloma (APOLLO): an open-label, randomised, phase 3 trial. Lancet Oncol 2021; 22: 801–812. [DOI] [PubMed] [Google Scholar]
- 19. Lonial S, Weiss BM, Usmani SZ, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 2016; 387: 1551–1560. [DOI] [PubMed] [Google Scholar]
- 20. Martin T, Strickland S, Glenn M, et al. Phase I trial of isatuximab monotherapy in the treatment of refractory multiple myeloma. Blood Cancer J 2019; 9: 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Moreau P, Dimopoulos MA, Mikhael J, et al. Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): a multicentre, open-label, randomised phase 3 trial. Lancet 2021; 397: 2361–2371. [DOI] [PubMed] [Google Scholar]
- 22. Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood 2020; 136: 936–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Attardi E, Pilerci S, Attucci I, et al. Ninety-minute daratumumab infusions for relapsed and refractory multiple myeloma: two years of Italian single-center observational study. Clin Lymphoma Myeloma Leuk 2021; 21: e850–e852. [DOI] [PubMed] [Google Scholar]
- 24. Barr H, Dempsey J, Waller A, et al. Ninety-minute daratumumab infusion is safe in multiple myeloma. Leukemia 2018; 32: 2495–2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Bonello F, Rocchi S, Barilà G, et al. Safety of rapid daratumumab infusion: a retrospective, multicenter, real-life analysis on 134 patients with multiple myeloma. Front Oncol 2022; 12: 851864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Gordan L, Chang M, Lafeuille MH, et al. Real-world utilization and safety of daratumumab IV rapid infusions administered in a community setting: a retrospective observational study. Drugs Real World Outcomes 2021; 8: 187–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Hamadeh IS, Reese ES, Arnall JR, et al. Safety and cost benefits of the rapid daratumumab infusion protocol. Clin Lymphoma Myeloma Leuk 2020; 20: 526–532. [DOI] [PubMed] [Google Scholar]
- 28. Lombardi J, Boulin M, Devaux M, et al. Safety of ninety-minute daratumumab infusion. J Oncol Pharm Pract 2021; 27: 1080–1085. [DOI] [PubMed] [Google Scholar]
- 29. Usmani SZ, Karanes C, Bensinger WI, et al. Final results of a phase 1b study of isatuximab short-duration fixed-volume infusion combination therapy for relapsed/refractory multiple myeloma. Leukemia 2021; 35: 3526–3533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Arnall JR, Moore DC, Hill HL, et al. Enhancing the feasibility of outpatient daratumumab administration via a split-dosing strategy with initial doses. Leuk Lymphoma 2019; 60: 2295–2298. [DOI] [PubMed] [Google Scholar]
- 31. Coffman K, Carstens C, Fajardo S. Daratumumab infusion reaction rates pre- and post-addition of montelukast to pre-medications. J Oncol Pharm Pract. Epub ahead of print 12 January 2022. DOI: 10.1177/10781552211072876. [DOI] [PubMed] [Google Scholar]
- 32. Hofmeister CC, Lonial S. How to integrate elotuzumab and daratumumab into therapy for multiple myeloma. J Clin Oncol 2016; 34: 4421–4430. [DOI] [PubMed] [Google Scholar]
- 33. Moore DC, Arnall JR, Thompson DL, et al. Evaluation of montelukast for the prevention of infusion-related reactions with daratumumab. Clin Lymphoma Myeloma Leuk 2020; 20: e777–e781. [DOI] [PubMed] [Google Scholar]
- 34. Nahi H, Usmani SZ, Mateos M-V, et al. Subcutaneous daratumumab with rapid corticosteroid tapering in relapsed or refractory multiple myeloma patients: part 3 update of the open-label, multicenter, phase 1b PAVO study. Blood 2021; 138: 1667. [DOI] [PubMed] [Google Scholar]
- 35. Nooka AK, Gleason C, Sargeant MO, et al. Managing infusion reactions to new monoclonal antibodies in multiple myeloma: daratumumab and elotuzumab. J Oncol Pract 2018; 14: 414–422. [DOI] [PubMed] [Google Scholar]
- 36. Al Hadidi S, Miller-Chism CN, Kamble R, et al. Safety analysis of five randomized controlled studies of daratumumab in patients with multiple myeloma. Clin Lymphoma Myeloma Leuk 2020; 20: e579–e589. [DOI] [PubMed] [Google Scholar]
- 37. Dimopoulos MA, Oriol A, Nahi H, et al. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med 2016; 375: 1319–1331. [DOI] [PubMed] [Google Scholar]
- 38. Lonial S, Dimopoulos M, Palumbo A, et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. N Engl J Med 2015; 373: 621–631. [DOI] [PubMed] [Google Scholar]
- 39. Lonial S, Vij R, Harousseau JL, et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. J Clin Oncol 2012; 30: 1953–1959. [DOI] [PubMed] [Google Scholar]
- 40. Mikhael J, Belhadj-Merzoug K, Hulin C, et al. A phase 2 study of isatuximab monotherapy in patients with multiple myeloma who are refractory to daratumumab. Blood Cancer J 2021; 11: 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Luo MM, Usmani SZ, Mateos MV, et al. Exposure-response and population pharmacokinetic analyses of a novel subcutaneous formulation of daratumumab administered to multiple myeloma patients. J Clin Pharmacol 2021; 61: 614–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lancman G, Sastow D, Aslanova M, et al. Effect of intravenous immunoglobulin on infections in multiple myeloma (MM) patients receiving daratumumab. Blood 2020; 136: 6–7.32614958 [Google Scholar]
- 43. Burns EA, Ensor JE, Anand K, et al. Opportunistic infections in patients receiving daratumumab regimens for multiple myeloma (MM). Blood 2021; 138: 4740. [Google Scholar]
- 44. Frerichs KA, Bosman PWC, Nijhof IS, et al. Cytomegalovirus reactivation in a patient with extensively pretreated multiple myeloma during daratumumab treatment. Clin Lymphoma Myeloma Leuk 2019; 19: e9–e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Lavi N, Okasha D, Sabo E, et al. Severe cytomegalovirus enterocolitis developing following daratumumab exposure in three patients with multiple myeloma. Eur J Haematol. Epub ahead of print 18 August 2018. DOI: 10.1111/ejh.13164. [DOI] [PubMed] [Google Scholar]
- 46. Nahi H, Chrobok M, Gran C, et al. Infectious complications and NK cell depletion following daratumumab treatment of multiple myeloma. PLoS ONE 2019; 14: e0211927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Nakagawa R, Onishi Y, Kawajiri A, et al. Preemptive therapy for cytomegalovirus reactivation after daratumumab-containing treatment in patients with relapsed and refractory multiple myeloma. Ann Hematol 2019; 98: 1999–2001. [DOI] [PubMed] [Google Scholar]
- 48. Sato S, Kambe E, Tamai Y. Disseminated cryptococcosis in a patient with multiple myeloma treated with daratumumab, lenalidomide, and dexamethasone. Intern Med 2019; 58: 843–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Tabata R, Sato N, Yamauchi N, et al. Cytomegalovirus reactivation in patients with multiple myeloma administered daratumumab-combination regimens. Ann Hematol 2022; 101: 465–467. [DOI] [PubMed] [Google Scholar]
- 50. Ueno T, Ohta T, Imanaga H, et al. Listeria monocytogenes bacteremia during isatuximab therapy in a patient with multiple myeloma. Intern Med 2021; 60: 3605–3608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Cottini F, Huang Y, Williams N, et al. Real world experience of daratumumab: evaluating lymphopenia and adverse events in multiple myeloma patients. Front Oncol 2020; 10: 575168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. National Institute of Diabetes and Digestive and Kidney Diseases. Livertox: clinical and research information on drug-induced liver injury. Bethesda, MD: National Institute of Diabetes and Digestive and Kidney Diseases, 2012. [Google Scholar]
- 53. Kikuchi T, Kusumoto S, Tanaka Y, et al. Hepatitis B virus reactivation in a myeloma patient with resolved infection who received daratumumab-containing salvage chemotherapy. J Clin Exp Hematop 2020; 60: 51–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Mustafayev K, Torres H. Hepatitis B virus and hepatitis C virus reactivation in cancer patients receiving novel anticancer therapies. Clin Microbiol Infect. Epub ahead of print March 2022. DOI: 10.1016/j.cmi.2022.02.042. [DOI] [PubMed] [Google Scholar]
- 55. National Comprehensive Cancer Network. Prevention and treatment of cancer related infections (Version 1.2021), 2021. https://www.nccn.org/professionals/physician_gls/pdf/infections.pdf.
- 56. Kumar SK, Callander NS, Adekola K, et al. Multiple myeloma, version 3.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2020; 18: 1685–1717. [DOI] [PubMed] [Google Scholar]
- 57. Chari A, Samur MK, Martinez-Lopez J, et al. Clinical features associated with COVID-19 outcome in multiple myeloma: first results from the international myeloma society data set. Blood 2020; 136: 3033–3040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Cook G, John Ashcroft A, Pratt G, et al. Real-world assessment of the clinical impact of symptomatic infection with severe acute respiratory syndrome coronavirus (COVID-19 disease) in patients with multiple myeloma receiving systemic anti-cancer therapy. Br J Haematol 2020; 190: e83–e86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Hultcrantz M, Richter J, Rosenbaum CA, et al. COVID-19 infections and clinical outcomes in patients with multiple myeloma in New York City: a cohort study from five academic centers. Blood Cancer Discov 2020; 1: 234–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tixagevimab and cilgavimab (Evusheld) for pre-exposure prophylaxis of COVID-19. JAMA 2022; 327: 384–385. [DOI] [PubMed] [Google Scholar]
- 61. Hsi ED, Steinle R, Balasa B, et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res 2008; 14: 2775–2784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Eleutherakis-Papaiakovou E, Gavriatopoulou M, Ntanasis-Stathopoulos I, et al. Elotuzumab in combination with pomalidomide and dexamethasone for the treatment of multiple myeloma. Expert Rev Anticancer Ther 2019; 19: 921–928. [DOI] [PubMed] [Google Scholar]
- 63. Lonial S, Lee HC, Badros A, et al. Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study. Lancet Oncol 2020; 21: 207–221. [DOI] [PubMed] [Google Scholar]
- 64. Lonial S, Lee HC, Badros A, et al. Longer term outcomes with single-agent belantamab mafodotin in patients with relapsed or refractory multiple myeloma: 13-month follow-up from the pivotal DREAMM-2 study. Cancer 2021; 127: 4198–4212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Farooq AV, Degli Esposti S, Popat R, et al. Corneal epithelial findings in patients with multiple myeloma treated with antibody-drug conjugate belantamab mafodotin in the pivotal, randomized, DREAMM-2 study. Ophthalmol Ther 2020; 9: 889–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Cohen AD, Lee HC, Trudel S, et al. MM-250: impact of prolonged dose delays on response with belantamab mafodotin (belamaf; GSK2857916) treatment in the DREAMM-2 study: 13-month follow-up. Clinical Lymphoma Myeloma and Leukemia 2020; 20: S304–S305. [Google Scholar]
- 67. Hultcrantz M, Kleinman D, Ghataorhe P, et al. Exploring alternative dosing regimens of single-agent belantamab mafodotin on safety and efficacy in patients with relapsed or refractory multiple myeloma: DREAMM-14. Blood 2021; 138: 1645–1645.34734998 [Google Scholar]
- 68. Lonial S, Nooka AK, Thulasi P, et al. Management of belantamab mafodotin-associated corneal events in patients with relapsed or refractory multiple myeloma (RRMM). Blood Cancer J 2021; 11: 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Munshi NC, Anderson LD, Jr, Shah N, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med 2021; 384: 705–716. [DOI] [PubMed] [Google Scholar]
- 70. Berdeja JG, Madduri D, Usmani SZ, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet 2021; 398: 314–324. [DOI] [PubMed] [Google Scholar]
- 71. Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant 2019; 25: 625–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T–cell therapy – assessment and management of toxicities. Nat Rev Clin Oncol 2018; 15: 47–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Santomasso BD, Park JH, Salloum D, et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov 2018; 8: 958–971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Xiao X, Huang S, Chen S, et al. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. J Exp Clin Cancer Res 2021; 40: 367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Cohen AD, Parekh S, Santomasso BD, et al. Incidence and management of car-T neurotoxicity in patients with multiple myeloma treated with ciltacabtagene autoleucel in CARTITUDE studies. Blood Cancer J 2022; 12: 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018; 378: 439–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Nahas GR, Komanduri KV, Pereira D, et al. Incidence and risk factors associated with a syndrome of persistent cytopenias after CAR-T cell therapy (PCTT). Leuk Lymphoma 2020; 61: 940–943. [DOI] [PubMed] [Google Scholar]
- 78. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017; 377: 2531–2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Schaefer A, Saygin C, Maakaron J, et al. Cytopenias after chimeric antigen receptor T-cells (CAR-T) infusion; patterns and outcomes. Biol Blood Marrow Transplant 2019; 25: S171. [Google Scholar]
- 80. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2019; 380: 45–56. [DOI] [PubMed] [Google Scholar]
- 81. Tai YT, Landesman Y, Acharya C, et al. CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications. Leukemia 2014; 28: 155–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Tan DS, Bedard PL, Kuruvilla J, et al. Promising SINES for embargoing nuclear-cytoplasmic export as an anticancer strategy. Cancer Discov 2014; 4: 527–537. [DOI] [PubMed] [Google Scholar]
- 83. Chari A, Vogl DT, Gavriatopoulou M, et al. Oral selinexor-dexamethasone for triple-class refractory multiple myeloma. N Engl J Med 2019; 381: 727–738. [DOI] [PubMed] [Google Scholar]
- 84. Grosicki S, Simonova M, Spicka I, et al. Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): a randomised, open-label, phase 3 trial. Lancet 2020; 396: 1563–1573. [DOI] [PubMed] [Google Scholar]
- 85. Tao Y, Zhou H, Niu T. Safety and efficacy analysis of selinexor-based treatment in multiple myeloma, a meta-analysis based on prospective clinical trials. Front Pharmacol 2021; 12: 758992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Chen CI, Bahlis NJ, Gasparetto C, et al. Selinexor in combination with pomalidomide and dexamethasone (SPd) for treatment of patients with relapsed refractory multiple myeloma (RRMM). Blood 2020; 136: 18–19. [Google Scholar]
- 87. Gasparetto C, Lentzsch S, Schiller GJ, et al. Selinexor, daratumumab, and dexamethasone in patients with relapsed/refractory multiple myeloma (MM). J Clin Oncol 2020; 38: 8510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Gasparetto C, Schiller GJ, Tuchman SA, et al. Once weekly selinexor, carfilzomib and dexamethasone in carfilzomib non-refractory multiple myeloma patients. Br J Cancer 2022; 126: 718–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Jakubowiak AJ, Jasielec JK, Rosenbaum CA, et al. Phase 1 study of selinexor plus carfilzomib and dexamethasone for the treatment of relapsed/refractory multiple myeloma. Br J Haematol 2019; 186: 549–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Nooka AK, Costa LJ, Gasparetto CJ, et al. Guidance for use and dosing of selinexor in multiple myeloma in 2021: consensus from international myeloma foundation expert roundtable. Clin Lymphoma Myeloma Leuk. Epub ahead of print February 2022. DOI: 10.1016/j.clml.2022.01.014. [DOI] [PubMed] [Google Scholar]
- 91. Harrison SJ, Minnema MC, Lee HC, et al. A phase 1 first in human (FIH) study of AMG 701, an anti-B-cell maturation antigen (BCMA) half-life extended (HLE) BiTE® (bispecific T-cell engager) molecule, in relapsed/refractory (RR) multiple myeloma (MM). Blood 2020; 136: 28–29. [Google Scholar]
- 92. Usmani SZ, Garfall AL, Van De Donk N, et al. Teclistamab, a B-cell maturation antigen × CD3 bispecific antibody, in patients with relapsed or refractory multiple myeloma (MajesTEC-1): a multicentre, open-label, single-arm, phase 1 study. Lancet 2021; 398: 665–674. [DOI] [PubMed] [Google Scholar]
- 93. Sebag M, Raje NS, Bahlis NJ, et al. Elranatamab (PF-06863135), a B-cell maturation antigen (BCMA) targeted CD3-engaging bispecific molecule, for patients with relapsed or refractory multiple myeloma: results from magnetismm-1. Blood 2021; 138: 895. [Google Scholar]
- 94. Kumar S, D’Souza A, Shah N, et al. A phase 1 first-in-human study of Tnb-383B, a BCMA × CD3 bispecific T-cell redirecting antibody, in patients with relapsed/refractory multiple myeloma. Blood 2021; 138: 900. [Google Scholar]
- 95. Zonder JA, Richter J, Bumma N, et al. Early, deep, and durable responses, and low rates of cytokine release syndrome with REGN5458, a BCMAxCD3 bispecific monoclonal antibody, in a phase 1/2 first-in-human study in patients with relapsed/refractory multiple myeloma (RRMM). Blood 2021; 138: 160.33831168 [Google Scholar]
- 96. Krishnan AY, Minnema MC, Berdeja JG, et al. Updated phase 1 results from MonumenTAL-1: first-in-human study of talquetamab, a G protein-coupled receptor family C group 5 member D × CD3 bispecific antibody, in patients with relapsed/refractory multiple myeloma. Blood 2021; 138: 158–158. [Google Scholar]
- 97. Krishnan AY, Garfall AL, Mateos M-V, et al. Updated phase 1 results of teclistamab, a B-cell maturation antigen (BCMA) × CD3 bispecific antibody, in relapsed/refractory multiple myeloma (MM). J Clin Oncol 2021; 39: 8007. [Google Scholar]
- 98. Moreau P, Usmani SZ, Garfall AL, et al. Updated results from MajesTEC-1: phase 1/2 study of teclistamab, a B-Cell maturation antigen X CD3 bispecific antibody, in relapsed/refractory multiple myeloma. Blood 2021; 138: 896–896. [Google Scholar]
- 99. Chauhan D, Velankar M, Brahmandam M, et al. A novel Bcl-2/Bcl-X(L)/Bcl-W inhibitor ABT-737 as therapy in multiple myeloma. Oncogene 2007; 26: 2374–2380. [DOI] [PubMed] [Google Scholar]
- 100. Bahlis NJ, Baz R, Harrison SJ, et al. Phase I study of venetoclax plus daratumumab and dexamethasone, with or without bortezomib, in patients with relapsed or refractory multiple myeloma with and without T(11;14). J Clin Oncol 2021; 39: 3602–3612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Boccon-Gibod C, Talbot A, Le Bras F, et al. Carfilzomib, venetoclax and dexamethasone for relapsed/refractory multiple myeloma. Br J Haematol 2020; 189: e73–e76. [DOI] [PubMed] [Google Scholar]
- 102. Costa LJ, Davies FE, Monohan GP, et al. Phase 2 study of venetoclax plus carfilzomib and dexamethasone in patients with relapsed/refractory multiple myeloma. Blood Adv 2021; 5: 3748–3759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Kaufman JL, Gasparetto C, Schjesvold FH, et al. Targeting Bcl-2 with venetoclax and dexamethasone in patients with relapsed/refractory T(11;14) multiple myeloma. Am J Hematol 2021; 96: 418–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Kumar SK, Harrison SJ, Cavo M, et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol 2020; 21: 1630–1642. [DOI] [PubMed] [Google Scholar]