ASCO 2023: Hematologic Malignancies

CME

Key Studies in Hematologic Malignancies: Independent Conference Coverage of the 2023 ASCO Annual Meeting

Physicians: Maximum of 1.50 AMA PRA Category 1 Credits

Released: August 25, 2023

Expiration: August 24, 2024

Shaji K. Kumar
Shaji K. Kumar, MD
Jeffrey P. Sharman
Jeffrey P. Sharman, MD
Eunice S. Wang
Eunice S. Wang, MD

Activity

Progress
1
Course Completed

CAR T-Cell Therapies in MM

Shaji Kumar, MD:
BCMA is expressed on MM tumor cells and some late memory B-cells and plasma cells. Currently, 2 classes of BCMA-targeted therapies are approved to treat patients with R/R MM and provide a much-needed treatment alternative for this patient population. The CAR T-cell immunotherapies idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel), target BCMA and are currently approved by the FDA for patients with R/R MM after ≥4 prior lines of therapy including a PI, an IMiD, and an anti-CD38 Ab.33,34 Similarly, the BCMA-targeted bispecific antibodies teclistamab and elranatamab also are approved by the FDA for patients with R/R MM after ≥4 prior lines of therapy including a PI, an IMiD, and an anti-CD38 Ab.35, 36 

Efficacy of ide-cel was demonstrated in the phase II KarMMa trial, which enrolled patients who had received ≥3 previous lines of therapy including a PI, an IMiD, and an anti-CD38 Ab, with an ORR of 73% and a median PFS of 8.8 months.37 The phase II CARTITUDE-1 trial of cilta-cel in patients who had received ≥3 previous lines of therapy including a PI, an IMiD, and an anti-CD38 Ab demonstrated an ORR of 97.9%, with the median PFS not yet reached at 27.7 months of follow-up.38 

Two randomized phase III trials have compared these CAR T-cell therapies with SoC therapy in patients with R/R MM in earlier lines of treatment. In the recently published KarMMa-3 trial, patients with R/R MM after 2-4 prior lines of therapy including an IMiD, a PI, and an anti-CD38 Ab were randomized to receive ide-cel or standard combination regimens. Treatment with ide-cel resulted in longer PFS (13.3 months vs 4.4 months; HR: 0.49: 95% CI: 0.38-0.65; P <.001) than standard combination regimens.39 

At ASCO 2023, results were presented from the second of these trials, CARTITUDE-4, an open-label trial comparing cilta-cel with SoC in patients with MM who had received 1-3 prior lines of therapy.40,41 In addition, updated data from CARTITUDE-1 with long-term follow-up were presented.42

CARTITUDE-4: Study Design

CARTITUDE-4 was a randomized, open-label phase III trial assessing treatment with cilta-cel vs the SoC regimens pomalidomide/bortezomib/dexamethasone or daratumumab/pomalidomide/dexamethasone.40,41 Patients with R/R MM whose disease was lenalidomide refractory after 1-3 previous lines of therapy were enrolled, and patients’ prior therapies must include a PI and an IMiD. Patients who received prior BCMA‑targeting therapy were excluded. In total, 419 patients were randomly assigned to receive a cilta‑cel infusion or SoC administered until disease progression. 

The primary endpoint was PFS. Secondary endpoints included CR or better, overall ORR, MRD negativity, OS, safety, and patient-reported outcomes. 

Patients were stratified by the type of SoC therapy, ISS stage, and the number of previous lines of therapy. The current analysis was completed after a median follow-up of 15.9 months.

CARTITUDE-4: Baseline Characteristics

The patient groups were fairly balanced between the 2 treatment arms.40,41 Patients had a reasonably good PS, with 99.5% of patients having a PS of 1 or better in each arm. Approximately 94% of patients had ISS stage I or II disease in each arm. Of interest, approximately one fifth of patients had soft tissue plasmacytomas.

Overall, a high proportion of the trial population had higher-risk disease, suggesting that patients may have progressed after their initial lines of therapy relatively quickly. The median time from diagnosis was approximately 3 years in each arm, and approximately one third of patients had received only 1 prior line of therapy. Approximately 26% of patients were triple-class exposed, 15% were triple-class refractory, and 22% previously had received daratumumab.

CARTITUDE-4: PFS (ITT Population)

Cilta-cel demonstrated a clear improvement in PFS in the intention-to-treat (ITT) population compared with SoC (median PFS: not reached vs 11.8 months; HR: 0.26; 95% CI: 0.18-0.38; P <.0001).40,41 The median PFS of approximately 12 months reported in the SoC control arm is what one would have anticipated with these 2 triplet regimens in patients who were lenalidomide refractory.  

The 1-year PFS rate was 76% with cilta-cel and 49% with SoC. Of interest, a higher proportion of patients had early PFS events in the cilta‑cel arm during and directly after the bridging phase prior to cilta-cel infusion, which then caught up over time. These data likely reflect the time taken to initiate apheresis and cilta-cel infusion.

CARTITUDE-4: PFS in Key Subgroups

Subgroup analyses demonstrated a clear PFS benefit with CAR T-cell therapy regardless of the number of lines of therapy, but patients with 2‑3 prior lines of therapy may have had a slightly greater relative benefit compared with patients who had received only 1 prior line of therapy.40,41  

The PFS benefit also was seen across ISS stages, but fewer patients had ISS stage III disease. The presence of soft tissue plasmacytomas seems to have somewhat affected outcomes, but these patients still benefited from the use of CAR T‑cell therapy.

Tumor burden and prior exposure to anti-CD38 therapy did not seem to have an impact on the benefit of cilta-cel. The PFS benefit also appeared to be similar regardless of cytogenetic risk, but there may have been a greater benefit in the high‑risk patient population. 

CARTITUDE-4: Key Secondary Endpoints (ITT)

In the ITT analysis, ORR with cilta-cel was 84.6% (58.2% stringent CR [sCR]) vs 67.3% (15.2% sCR) with SoC.40,41 The median DoR was not reached with cilta-cel and 16.6 months with SoC, whereas the 12-month DoR rates were 84.7% and 63.0%, respectively. Rates of MRD negativity were 60.6% with cilta-cel and 15.6% with SoC (odds ratio: 8.7; P <.0001). 

OS data were immature at the time of analysis; there were 39 deaths in the cilta-cel arm and 47 in the control arm (HR: 0.78; 95% CI: 0.5-1.2; P = .26).

CARTITUDE-4: Efficacy Results in As-Treated Population

In the as-treated population of patients who received cilta-cel as study treatment, cilta-cel was associated with an ORR of 99.4%, including 68.8%of patients with sCR, and an MRD negativity rate (at a sensitivity of 10-5) of 72% among 176 assessed patients.40,41 

At 12 months from the time of apheresis, 90% of patients were alive and progression free. The 12-month PFS rate from the time of cilta-cel infusion was 85%. In comparison, the 12-month PFS rate reported in the CARTITUDE-1 trial in a more heavily pretreated population was 76%, and the rate of MRD negativity in this population was 58%.43 

Of note, 32 patients included in the ITT population did not receive cilta-cel as study treatment due to disease progression or death during bridging therapy and/or lymphodepletion and were not included in the as-treated analysis, but 20 of these patients received cilta-cel as subsequent therapy.

CARTITUDE-4: TEAEs

Rates of any TEAEs and serious TEAEs were similar between arms. Serious TEAEs of any grade occurred in 44.2% of patients in the cilta‑cel arm and 38.9% of patients in the SoC arm.40,41  

Rates of hematologic toxicities with cilta-cel were as one would anticipate based on prior results. Most patients experienced neutropenia, and rates of grade 3/4 neutropenia were 89.9% with cilta-cel vs 82.2% with SoC. Rates of grade 3/4 thrombocytopenia were 41.3% and 18.8%, respectively, and rates of grade 3/4 lymphopenia were 20.7% and 12.0%, respectively.  

Infections were common in both treatment arms, with grade 3 or 4 infections occurring in 26.9% of patients in the cilta‑cel arm and 24.5% of patients in the SoC arm. In addition, 9 (4.3%) second primary malignancies were reported in the cilta-cel arm and 14 (6.7%) in the SoC arm. This is important to note when we talk about the long‑term follow-up of the CARTITUDE‑1 trial. 

Overall, there were 10 deaths due to TEAEs in the cilta‑cel arm, predominantly from COVID‑19 or other infections. There were 5 deaths due to TEAEs in the SoC arm, again predominantly from infections, but 1 patient died from progressive multifocal leukoencephalopathy and 1 patient from pulmonary embolism.

CARTITUDE-4: CRS and Neurotoxicity Associated With Cilta-Cel

CRS and neurotoxicity are of particular interest with CAR T‑cell therapy and other immunotherapies. The any-grade CRS rate was 76.1% in CARTITUDE-4, and only 2 patients (1%) developed grade 3/4 CRS, which is substantially lower than what was noted in the initial studies of cilta‑cel.40,41,43 The median time to onset of CRS was 8 days, and the median duration was 3 days. 

ICANS was observed in 4.5% of patients, with no grade 3 or 4 events reported. This incidence was lower than that reported in the initial phase I/II study of cilta-cel.40,41,43 The median time to onset of ICANS was 10 days, and the median duration was 2 days. 

Of interest, 9.1% of patients developed cranial nerve palsy in this trial, and this is important to keep in mind for patients with R/R MM who are receiving CAR T‑cell therapy. Peripheral neuropathy and other neurologic toxicities were uncommon, and there were no fatal neurotoxicities. 

CARTITUDE-4: Clinical Implications

In this analysis from CARTITUDE-4, cilta-cel was associated with superior PFS compared with SoC therapy in patients with lenalidomide-refractory MM after 1-3 prior lines of therapy.40,41 However, these data do not explore whether treatment with cilta-cel in earlier lines of therapy improves outcomes compared with CAR T-cell therapy in later lines of treatment. When comparing trials in earlier vs later lines of therapy, the 12-month PFS rate from CARTITUDE-4 was 76%, and the 12-month PFS rate from CARTITUDE-1 was 77%.40,43 The KarMMa-3 trial reported a median PFS of 13.3 months with ide-cel, and KarMMa reported a median PFS of 8.8 months.37,39 

This raises an important question regarding the optimal sequencing of therapies. Given that the duration of benefit from CAR T-cell therapy may be similar whether used in earlier or later relapse—and that there are AEs associated with CAR T-cell therapy that we are concerned about—should we reserve CAR T-cell therapy for later lines of treatment? At this point, CAR T-cell therapy remains an option after ≥4 prior lines of therapy.

The CARTITUDE-4 phase III trial evaluated ciltacabtagene autoleucel (cilta-cel) vs pomalidomide/bortezomib/dexamethasone or daratumumab/pomalidomide/dexamethasone (standard of care [SoC]) in patients with lenalidomide-refractory MM after 1-3 prior lines of therapy. In which of the following patient populations was progression-free survival (PFS) benefit demonstrated with cilta-cel compared with SoC?

CARTITUDE-1 Update: Study Design

Also at ASCO 2023, the end-of-study analysis of the phase Ib/II CARTITUDE-1 trial, which led to the FDA approval of cilta-cel in patients with R/R MM after ≥4 prior therapies including a PI, an IMiD, and an anti-CD38 Ab, was presented.34,42 This single‑arm, open‑label trial enrolled patients with R/R MM with measurable disease after ≥3 prior therapies including a PI, an IMiD, and an anti-CD38 Ab or who were double refractory to a PI and an IMiD. 

Patients were enrolled, underwent leukapheresis, received bridging chemotherapy as required clinically, and then underwent lymphodepletion with fludarabine/cyclophosphamide followed by infusion of the CAR T-cells. The primary endpoint for CARTITUDE‑1 was safety for phase I and ORR for phase II. Secondary endpoints included PFS, OS, and MRD negativity (at a sensitivity of 10-5). At 27.7 months of follow-up, cilta-cel in patients who were heavily pretreated demonstrated an ORR of 97.9% with median PFS not yet reached.38 The current analysis reports end-of-study findings at a median follow-up of 33.4 months.

CARTITUDE-1 Update: Efficacy Summary

This update confirmed that cilta-cel was associated with an ORR of 97.9% (82.5% sCR).42 The median DOR and PFS were quite impressive at nearly 3 years for each (33.9 months and 34.9 months, respectively). The median OS was not reached, but the 3-year OS rate was 62.9%. 

Sustained MRD negativity for ≥12 months was attained in more than 50% of evaluable patients (26 of 49); 18 of those 26 patients remained MRD negative with a CR or better at 24 months post infusion.

CARTITUDE-1 Update: PFS by CR and MRD Negativity

The median PFS with cilta-cel was 34.9 months in the overall population, with a 3-year PFS rate of 47.5%. Among patients who achieved a CR or better at any time during the study, the median PFS increased to 38.2 months, with a 3-year PFS rate of 59.8%. Among patients with MRD negativity sustained for ≥12 months, the median PFS was not reached, and the 30-month PFS rate was 74.9%.42 Clearly, patients who had a deep or sustained response had a significant PFS benefit.

CARTITUDE-1 Update: Study Deaths and Safety

Regarding safety in CARTITUDE‑1, a few additional pieces of information have become evident with longer‑term follow-up.42 At a median follow-up of 27.2 months, 35 deaths have occurred among the 97 enrolled patients. Approximately one half of these deaths (18 of 35) were due to causes other than MM disease progression, and 17 were due to progressive disease. 

The most common cause of death other than progressive disease was infections, including pneumonia (n = 2 unrelated to treatment), respiratory failure (n = 3 unrelated to treatment and n = 1 related to treatment), and sepsis (n = 2 unrelated to treatment and n = 2 related to treatment). There were 3 deaths due to AML and 1 death due to MDS. One patient with AML had MDS and cytogenetic abnormalities prior to their cilta‑cel infusion. The cytogenetic profile details are not available for the others. There were 5 additional deaths since the last analysis, but these all were considered unrelated to cilta-cel. 

It is important to note the spectrum of AEs that have occurred among patients receiving cilta-cel, particularly the impact of infections in this patient population and the potential signal of second primary malignancies. It is unknown if the emergence of these second primary malignancies is related to the fact that these patients have lived longer than what they otherwise would have, representing a natural history of the disease, or whether these malignancies are related to the perturbation of their immune system or immune surveillance, allowing cancers that might have been present even before to progress more rapidly. Certainly, we have a lot to learn about those aspects.

CARTITUDE-1 Clinical Implications

This end-of-study update from CARTITUDE-1 demonstrated that a single dose of cilta-cel was associated with deep and durable responses for patients who were heavily pretreated with R/R MM and provides a lot of interesting data. This follow-up clearly confirms this initial observation about the safety and efficacy of cilta-cel. The median PFS of nearly 3 years in a heavily pretreated patient population is outstanding. The investigators reported that patients will continue to be followed up for survival and safety in the 15-year CARTINUE trial. As previously noted, questions remain about the optimal sequencing of CAR T-cell therapies and if the occurrences of second primary malignancies are therapy related.

MajesTEC-1 Update: Study Design

In addition to CAR T-cell therapy, bispecific antibodies were another type of immunotherapy in MM for which we saw important updates at ASCO 2023. These agents are engineered artificial antibodies that bind to the CD3 subunit within the T-cell receptor with one arm and to a tumor-associated antigen with the other.44 

Teclistamab is an off-the-shelf anti-BCMA x CD3 bispecific antibody that is approved for patients with R/R MM who have received ≥4 prior lines of therapy including a PI, an IMiD, and anti-CD38 Ab.35 This approval was based on results from the phase I/II MajesTEC-1 trial, which evaluated the safety and efficacy of teclistamab in patients with triple class‒exposed R/R MM with no prior BCMA therapy.45 The trial population had similar characteristics as the population from the CARTITUDE‑1 study. 

At ASCO 2023, van de Donk and colleagues46 presented an update from MajesTEC-1 after a median follow-up of 23 months. The phase I portion included IV and SC dose escalation to find the randomized phase II dosing. Then, in the phase II dose-expansion cohort, patients received teclistamab at 1.5 mg/kg SC weekly, which is the approved dose for clinical use. Responding patients could switch to every-2-week or every-4-week dosing after 6 months of therapy. The primary endpoint for MajesTEC‑1 was ORR. Key secondary endpoints included pharmacokinetics/pharmacodynamics, DoR, PFS, OS, uMRD, safety, and health-related quality of life. After a median follow-up of 14.1 months, an ORR of 63% was reported with teclistamab.45

MajesTEC-1 Update: Baseline Characteristics

The baseline characteristics of patients enrolled on this trial were as expected.46 The median age of enrolled patients was 64 years (range: 33-84), 58.2% of patients were male, 81.2% of patients were White, and 12.7% of patients were Black. In this population, 17% of patients had extramedullary disease, 25.7% had high-risk cytogenetics, and 87.7% had ISS stage I-II disease. The median time since diagnosis was 6 years, with a median of 5 previous lines of therapy; 70.3% of patients were penta exposed, and 30.3% were penta refractory. After a median follow-up of 23 months, 47 of 165 patients remained on treatment, with 42 patients receiving an every-2-week regimen and 9 patients receiving an every-4-week regimen.

MajesTEC-1 Update: Response and DoR

With longer-term follow-up, response rates have remained quite consistent, with an ORR of 63.0%, including 45.5% of patients achieving a CR or better.46 The ORR was higher among patients who had received ≤3 prior lines of therapy compared with those who had received >3 prior lines of therapy (74.4% vs 59.0%, respectively). The ORR in patients with high‑risk cytogenetics and/or extramedullary disease was slightly lower at 53.3%. 

Among patients who responded to teclistamab therapy, the responses were durable, with a median DoR of 21.6 months in all responding patients and 26.7 months among patients with a CR or better. The median time to achieve a CR or better was 4.6 months (range: 1.6-18.5).

MajesTEC-1 Update: Efficacy Summary

The median PFS was 11 months for the overall population and 27 months for the 45.5% of patients with a CR or better. The median OS was 22 months for the overall population and was not reached for patients with a CR or better.  

Among 42 patients evaluable for MRD at Day 100, the rate of uMRD at a 10-5 sensitivity was 81%. Among 54 patients evaluable at any point, the rate of uMRD was 81.5%.46 These results suggest that teclistamab is inducing deep responses in these patients with advanced R/R MM.

MajesTEC-1 Update: Safety

In this longer follow-up, 1 AE led to a dose reduction and 8 AEs led to discontinuations (5 due to infection).46 However, there were 7 treatment‑related deaths, including 4 due to COVID‑19, reflecting the period of enrollment during the pandemic and highlighting the increased susceptibility to infection in general. 

The most frequent hematologic AEs were neutropenia (71.5%; 65.5% grade 3/4), anemia (54.5%; 37.6% grade 3/4), thrombocytopenia (42.4%; 22.4% grade 3/4), and lymphopenia (36.4%; 34.5% grade 3/4). Although rates of neutropenia may have been a bit lower compared with rates reported with CAR T-cell therapy, neutropenia remains a predominant AE for these BCMA-targeted immunotherapies and is often grade 3 or 4. 

The most frequent nonhematologic AE was infection, occurring in 80% (55.2% grade 3/4) of patients. CRS was reported in 72% of patients, but only 1 (0.6%) patient experienced grade 3/4 CRS. 

ICANS occurred in 3% of patients with no grade 3/4 events. All cases of ICANS resolved. ICANS appears not to be as much of a concern here as we have seen with CAR T-cell therapy.  

Other common AEs included diarrhea (33.9%), pyrexia (31.5%), fatigue (29.1%), COVID-19 (29.1%), nausea (27.3%), cough (26.7%), injection-site erythema (26.1%), arthralgia (25.5%), headache (24.2%), constipation (21.8%), and hypogammaglobulinemia (20.6%).

MajesTEC-1 Update: Rate of New-Onset Grade ≥3 Infections

The rate of infections associated with BCMA-targeted CAR T-cell therapy and bispecific antibodies has been high, and current studies are assessing the various infection profiles with these classes of agents, including the type and kinetics of infections. 

There have been quite a few opportunistic infections, including Pneumocystis jirovecii pneumonia and others associated with teclistamab therapy. In the first year after starting treatment with teclistamab, approximately 20% of patients developed a new-onset grade ≥3 infection every quarter. Beyond 1 year, the proportion of new‑onset grade ≥3 infection seems to decrease over time. Factors that likely contribute to this decline include decreased dosing frequency, better management of infections and prophylaxis, better disease control long term, and patient selection. This is important information as we try to reduce the risk of infections in these patients.

MajesTEC-1 Update: Clinical Implications

In this update from MajesTEC-1 after a median follow-up of approximately 2 years, teclistamab has continued to provide deep and durable responses in patients with R/R MM, including in patients who have switched to a reduced dosing schedule. The reported ORR was 63%, with 45.5% of patients achieving a CR or better. In evaluable patients, 81.5% achieved uMRD (10-5) by Day 100. Rates of new grade ≥3 infections have decreased over time, and no new safety signals have been identified. 

The updated findings from the MajesTEC-1 trial with teclistamab appear to be consistent with earlier data in terms of both efficacy and toxicity. Teclistamab is the first BCMA-targeted bispecific antibody approved for R/R MM, and these data continue to show that this therapy offers an active, viable BCMA-directed therapy for our patients with heavily pretreated MM. However, continued questions remain about optimal strategies for toxicity prevention, monitoring, and management.

Elranatamab After Previous BCMA Therapy: Study Design

With the availability of multiple modalities to target BCMA for patients with MM, including the CAR T-cell therapies ide-cel and cilta-cel and the bispecific antibodies teclistamab and elranatamab, one of the big questions that arises is how to optimally sequence these different BCMA‑targeted therapies. 

Elranatamab is a novel humanized BCMA x CD3 bispecific antibody that was granted accelerated approved by the FDA in August 2023 for patients with R/R MM after ≥4 previous lines of therapy including a PI, an IMiD, and an anti-CD38 Ab. Results from cohort A of the phase II MagnetisMM-3 trial of elranatamab in patients with R/R MM who were BCMA therapy naive demonstrated an ORR of 61% at a median of 14.7 months of follow-up.47 At ASCO 2023, Nooka and colleagues48 presented a pooled analysis of 4 different MagnetisMM studies assessing the efficacy and safety of single-agent elranatamab in patients with R/R MM who have received ≥1 PI, ≥1 IMiD, and ≥1 anti-CD38 Ab and have been previously exposed to other BCMA-targeted therapy. The analysis included 87 patients with a median follow-up of 11.3 months.

Elranatamab After Previous BCMA Therapy: Baseline Characteristics

The median age in the cohort of patients who had received any prior BCMA therapy was 66 years, with 14.9% aged 75 years and older.48 Patients who received previous CAR T-cell therapy were slightly younger than those who received a BCMA-targeted ADC. 

High-risk cytogenetics were present in 24.1% of patients overall, 22% of patients who had received an ADC, and 30.6% of patients who had received CAR T-cell therapy. Approximately 23% of patients had stage III disease by the revised ISS criteria (27% with previous ADC therapy and 14% with previous CAR T-cell therapy), and 54% had extramedullary disease (56% with previous ADC and 47% with previous CAR T-cell therapy).

Elranatamab After Previous BCMA Therapy: Prior Treatments

Patients had received a median of 7 prior lines of therapy (range: 3-19).48 Prior BCMA therapy included an ADC for 59 patients (67.7%), CAR T-cell therapy for 36 patients (41.3%), and both therapies for 8 patients (9.2%). 

Most patients (85.1%) were penta exposed, and 55.2% were penta refractory. The proportion of patients with penta-refractory disease was 63.9% among those who had received CAR T-cell therapy and 54.2% among those who had received an ADC. The investigators defined penta refractory as prior exposure to 2 PIs, 2 IMiDs, and 1 anti-CD38 Ab.

Elranatamab After Previous BCMA Therapy: Efficacy Summary

The ORR for elranatamab in patients with any previous BCMA-targeted therapy was 46% (42.4% among patients with prior ADC therapy and 52.8% among patients with prior CAR T-cell therapy).48 The median DoR was reasonable at 17.1 months (13.6 months with previous ADC therapy and NE with previous CAR T-cell therapy), but the median PFS and OS were short. The median PFS was 5.5 months (3.9 months with previous ADC therapy and 10.0 months with previous CAR T-cell therapy), and the median OS was 12.1 months in the overall patient population and with previous ADC or CAR T-cell therapy.  

There seemed to be a numerical difference in these data depending on whether patients had previously received an ADC vs CAR T-cell therapy, but given the small number of patients in this pooled analysis, these data are difficult to interpret. 

Overall, the ORR is lower than the 61% ORR reported in the MagnetisMM-3 trial in patients with R/R MM who were BCMA therapy naive, highlighting a drop in the response rate among patients who have previously received BCMA‑targeted therapy.47 However, it is unclear how much of this reduction in efficacy is due to the more advanced disease state in these patients as we go through 2 different BCMA‑targeted therapies vs a clear loss of efficacy due to common mechanisms.

Elranatamab After Previous BCMA Therapy: Safety

The safety profile with elranatamab after prior BCMA therapy was consistent to what we have observed with elranatamab in general.48 We see hematologic toxicity including anemia (58.6%; 46.0% grade 3/4), neutropenia (44.8%; 41.4% grade 3/4), and thrombocytopenia (40.2%; 28.7% grade 3/4). In addition, CRS was reported in 65.5% of patients, but this was predominantly grade 1 or 2, with 2.3% developing grade 3/4 events. 

Infections were reported in 73.6% of patients, including 26.4% grade 3/4 events and 9.2% grade 5 events. The rates of infection with elranatamab after prior BCMA therapy observed in this analysis generally were comparable to those observed with other trials of BCMA‑targeted bispecific antibodies. 

There were 23 deaths related to TEAEs; these included disease progression (n = 11), COVID-19 (n = 5), septic shock or sepsis (n = 3), and other individual events. We hope that this rate of fatalities due to COVID-19 will be reduced moving forward, but it will be important to continue to monitor the risk of infection with this agent and BCMA-targeting therapies in general.

Elranatamab After Previous BCMA Therapy: CRS and ICANS

CRS developed in 65.5% of patients but was primarily grade 1 or 2, with only 2.3% of patients developing grade 3 CRS and no grade 4 events. The median time to onset of CRS was 2 days (range: 1-4), and the median time to resolution was also 2 days (range: 1-10). 

ICANS developed in 5 patients (5.7%), including 3 grade 2 events and 2 grade 3 events. The median time to onset of ICANS was 2 days (range: 1-3), as was the median time to ICANS resolution (range: 1-18).

Elranatamab Pooled Analysis: Clinical Implications

In this pooled analysis of patients with R/R MM who have received ≥1 PI, ≥1 IMiD, ≥1 anti-CD38 Ab, and prior BCMA-targeted therapy, treatment with the BCMA-targeting bispecific antibody elranatamab was associated with a confirmed ORR of 46%, a median PFS of 5.5 months, and a median OS of 12 months. The safety profile appeared to be consistent with data with elranatamab in the populations of patients who are BCMA therapy naive.47 

Overall, both CAR T-cell therapy and bispecific antibodies seem to be associated with an increased risk of infection that we need to consider when we think about the sequence in which these agents are being used and the stage at which they are used in patients with R/R MM. It is important to weigh the efficacy and safety of each treatment option when selecting the next therapy for patients with R/R MM. 

One advantage of CAR T-cell therapy is that it is a one-time infusion, after which patients are off treatment. This is a luxury that few patients with R/R MM have today. For patients who are eligible for both bispecific and CAR T-cell therapy, I may consider using CAR T‑cell therapy first and reserve bispecific antibody treatment for later. Unfortunately, because CAR T-cell therapy requires the patient to receive their infusion at larger treatment centers and each patient must wait for manufacturing of their own CAR T-cells, most patients in the community may ultimately receive a bispecific antibody before CAR T-cell therapy. 

In general, the optimal sequencing of bispecific antibodies vs CAR T-cell therapy remains a key question. Based on these and other data, there may a drop in the response rate of approximately 15% to 25% whether a patient receives CAR T-cell therapy after a bispecific antibody or a bispecific antibody after CAR T-cell therapy.48,49 However, these datasets have very small numbers of patients, and it is unclear whether the decline in response rate is related to the therapy itself or reflects the patients having gone through 1 additional line of therapy. 

With the potential for BCMA-targeted immunotherapy failure, additional targets for bispecific antibodies in MM are under investigation. For example, talquetamab binds to GPRC5D and cevostamab targets FcRH5 on MM cells and is currently being given as part of clinical trials for R/R MM.44 Talquetamab recently was granted accelerated approval by the FDA for adult patients with R/R MM who have received ≥4 prior lines of therapy including a PI, an IMiD, and an anti-CD38 Ab.50 

At ASCO 2023, very interesting but very early data were presented from a phase II trial with the combination of teclistamab and talquetamab for patients with R/R MM who were refractory or intolerant to established therapies (n = 27).51 The ORR in this heavily pretreated patient population with phase II dosing of teclistamab 3 mg/kg and talquetamab 0.8 mg/kg was 96.3%, including 40.7% of patients who achieved a CR or better. This therapy was not without AEs, and approximately 38% of patients in this dose group experienced grade 3/4 infection, with rates of cytopenias, CRS, and ICANS events similar to those reported in other trials of single-agent bispecific antibodies. Of interest, the presentation included data on patients with extramedullary disease, with an ORR of 85.7% and median PFS of 9.9 months. These data have led to a new trial exploring this combination specifically in patients with extramedullary disease, where treatment outcomes historically have been suboptimal.

Phase I PHE885 Study: Trial Design

Currently available CAR T-cell therapies are engineered by modifying a patient’s individual T-cells to express the CAR for personalized therapy.52 Because of this process, a challenge with CAR T-cell therapies is the extended manufacturing times required, which leads to reduced availability and delays in patient care. PHE885 is an investigational BCMA-directed CAR T-cell therapy that has a manufacturing time of <2 days and aims for a door-to-door time of <10 days in the United States.53,54 

At ASCO 2023, investigators presented updated results from a phase I study evaluating the safety and efficacy of PHE885 in patients with R/R MM following ≥2 prior lines of therapy including a PI, an IMiD, and an anti-CD38 Ab.55 The study enrolled 50 patients who underwent leukapheresis, lymphodepleting chemotherapy, and then a single infusion of PHE885. Escalating doses of PHE885 were studied, ranging from 2.5 x 106 cells to 20 x 106 cells. The primary endpoints were dose-limiting toxicities (DLTs) and safety. Secondary endpoints included the manufacturing success rate, ORR, and DoR. Results were presented after a median follow-up of 6.7 months.

Phase I PHE885 Study: Baseline Characteristics

The median age of enrolled patients was 65 years, (range: 45-81).55 Patients had received a median of 4 prior lines of therapy (range: 2-10). Extramedullary disease was present in 33% of patients, and 36% of patients had high-risk cytogenetic abnormalities. As reflected in the enrollment criteria, 94% of patients were triple refractory, and 62% were penta refractory. 

Bridging therapy was administered to 34% of patients in this clinical trial protocol.

Phase I PHE885 Study: DLTs and Deaths

DLTs developed in 8 of 50 patients and included neutropenia, neurotoxicity, transient ejection fraction decrease, atrial fibrillation, transaminitis, and increases in lipase and serum amylase.55 The ejection fraction decrease and atrial fibrillation were both in the context of CRS. 

There were 15 deaths on the study, including 13 due to disease progression, 1 due to hemorrhagic shock unrelated to treatment, and 1 due to lung infection in the context of PHE885-related myelosuppression.

Phase I PHE885 Study: Safety

Overall, 98% of patients had a TRAE of any grade, with 88% experiencing a TRAE of grade ≥3. The most frequent hematologic grade ≥3 TRAEs were neutropenia (80%), anemia (56%), thrombocytopenia (50%), and leukopenia (22%). Other nonhematologic grade ≥3 TRAEs included increased aspartate aminotransferase (16%) and decreased blood fibrinogen (12%).55

Phase I PHE885 Study: AEs of Special Interest

CRS of any grade occurred in 96% of patients receiving PHE885, and 10% of patients developed grade 3 CRS. The median time to onset of CRS was 8 days (range: 2-16), and the median duration of CRS was 4 days (range: 1-19). 

ICANS developed in 20% of patients, and 6% developed grade 3 ICANS. The median time to ICANS onset was 10.5 days (range: 6-16). All ICANS events occurred concomitant to CRS and resolved. There were no reports of parkinsonism or other delayed neurotoxicity.55

Phase I PHE885 Study: ORR

In this preliminary analysis, PHE885 was associated with an ORR of 98%, including 80% with a very good PR or better.55 When broken down by individual dose levels, the ORR was 100% at all but the lowest dose.

Phase I PHE885 Study: DoR

The median time to first response was approximately 1 month, and the median time to best response was 2.8 months. Investigators noted that many responses deepened over time, with conversions to CR or sCR occurring up to 18 months after the PHE885 infusion.55  

Across doses, MRD negativity was attained in 14 of 19 patients (74%) by Month 6.

Phase I PHE885 Study: Conclusions

Updated results from this phase I trial suggest that PHE885 may be a safe, effective therapy for patients with R/R MM after ≥2 lines of therapy. The manufacturing success rate was 100%, with short manufacturing times of approximately 2 days and <10 days to delivery for infusion. 

PHE885 was associated with an ORR of 100% at most doses evaluated, and AEs were as expected for BCMA-targeted CAR T-cell therapy.55  

An ongoing phase II trial (NCT05172596) is further evaluating PHE885 in patients with R/R MM.

Which of the following investigational BCMA-targeted CAR T-cell therapies has a manufacturing time of ≤2 days?

Summary

Treatment options targeting BCMA have transformed the care of patients with R/R MM. As we continue to learn more about these treatment approaches through ongoing and new clinical trials, we will be able to clarify when we should be using these agents in clinical practice, how we can best protect our patients from potential AEs associated with these immunotherapies (such as with infection prophylaxis or varying the dose to reduce rates of CRS), and whether we can combine or sequence BCMA-targeted therapy with standard antimyeloma backbone therapy or with even newer targeted approaches, such as those targeting GPRC5D or FcRH5. 

The availability of this expanding array of treatment choices for R/R MM ultimately means greater access to improve overall outcomes for more of our patients.