scholarly journals Pembrolizumab as Consolidation Strategy in Patients with Multiple Myeloma: Results of the GEM-Pembresid Clinical Trial

Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3615
Author(s):  
Noemí Puig ◽  
Luis A. Corchete-Sánchez ◽  
José J. Pérez-Morán ◽  
Julio Dávila ◽  
Teresa Paíno ◽  
...  

PD1 expression in CD4+ and CD8+ T cells is increased after treatment in multiple myeloma patients with persistent disease. The GEM-Pembresid trial analyzed the efficacy and safety of pembrolizumab as consolidation in patients achieving at least very good partial response but with persistent measurable disease after first- or second-line treatment. Moreover, the characteristics of the immune system were investigated to identify potential biomarkers of response to pembrolizumab. One out of the 17 evaluable patients showed a decrease in the amount of M-protein, although a potential late effect of high-dose melphalan could not be ruled out. Fourteen adverse events were considered related to pembrolizumab, two of which (G3 diarrhea and G2 pneumonitis) prompted treatment discontinuation and all resolving without sequelae. Interestingly, pembrolizumab induced a decrease in the percentage of NK cells at cycle 3, due to the reduction of the circulating and adaptive subsets (0.615 vs. 0.43, p = 0.007; 1.12 vs. 0.86, p = 0.02). In the early progressors, a significantly lower expression of PD1 in CD8+ effector memory T cells (MFI 1327 vs. 926, p = 0.03) was observed. In conclusion, pembrolizumab used as consolidation monotherapy shows an acceptable toxicity profile but did not improve responses in this MM patient population. The trial was registered at clinicaltrials.gov with identifier NCT02636010 and with EUDRACT number 2015-003359-23.

JCI Insight ◽  
2019 ◽  
Vol 4 (4) ◽  
Author(s):  
Alfred L. Garfall ◽  
Edward A. Stadtmauer ◽  
Wei-Ting Hwang ◽  
Simon F. Lacey ◽  
Jan Joseph Melenhorst ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 766-766 ◽  
Author(s):  
Aaron P. Rapoport ◽  
Edward A. Stadtmauer ◽  
Dan T. Vogl ◽  
Brendan M Weiss ◽  
Gwendolyn K. Binder-Scholl ◽  
...  

Abstract Background Despite recent therapeutic advances, multiple myeloma (MM) remains primarily an incurable cancer. Patients experiencing rapid recovery of T cells post autologous stem cell transplant (auto-SCT) may have improved outcomes, and spontaneous cellular responses to tumor can occur, suggesting immune mediated control of tumor is possible. We and others have investigated therapeutic cancer vaccines that have shown promise in pilot studies, in particular following post-transplant infusion of activated autologous T cells. However, efficacy of these approaches may be limited by thymic selection which restricts the repertoire of T cell receptors (TCRs) to low affinity TCRs that cannot recognize the low level of antigen present on most tumor cells. We hypothesized that incorporation of affinity-enhanced tumor antigen-specific TCRs into autologous T cells infused post-transplant would overcome this limitation and improve response rates in the post auto-SCT setting. Methods We report interim results of a Phase II clinical trial (NCT01352286) to evaluate the safety and activity of autologous T cells genetically engineered to express an affinity-enhanced TCR that recognizes the NY-ESO-1/LAGE-1 peptide complex HLA‐A*0201‐SLLMWITQC; these cells are infused in the setting of profound lymphodepletion that accompanies high dose chemotherapy administered during auto-SCT. Patients with high risk or relapsed MM, who are HLA‐A*0201 positive, and whose tumor is positive for NY-ESO-1 and/or LAGE-1 by RT-PCR are eligible. CD25 depleted CD4 and CD8 T cells are activated and expanded using anti-CD3/CD28 antibody conjugated microbeads, and genetically modified with a lentiviral vector containing the TCR construct at a multiplicity of infection of 1. Engineered T cells are administered four days after high dose melphalan and two days following auto-SCT, at a dose range of 1-10 billion total cells with a minimum gene modification requirement of 10%. Patients are evaluated for MM responses in accordance with the IMWG criteria at 6 weeks, and 3 and 6 months post infusion. At 3 months, patients start lenalidomide maintenance. The initial 6 patient phase is complete and a 20 patient extension phase is ongoing. Results Prior to enrollment on study, patients had received a median of 3 prior therapies including 6 with prior transplant. 50% of tumors contained high risk chromosomal abnormalities, and NY-ESO-1 expression is correlated with adverse prognosis. 20 patients (average age of 57) have been infused with an average of 2.3 X 109 engineered T cells (range 4.5 X 108-3.9 X 109); this reflects an average clinical scale transduction efficiency of 34% (range 18% – 49%). Infusions have been well tolerated, and the majority of adverse events were related to the high dose melphalan. Possibly related SAEs were neutropenia and thrombocytopenia, and GI and metabolism disorders including diarrhea, colitis, hyponatremia and hypomagnesemia. 10, 4, 2, and 2 patients have reached the 1 year, 9 month, 6 month and 3 month assessment timepoints, respectively, and 17/20 patients are alive. Best response by day 100 is sCR/CR in 2/15 (13%), nCR in 10/15 (67%), and PR in 3/15% (20%), which compares favorably to historic responses in patients undergoing first or second transplant. Engineered T cells expanded and persisted in blood and marrow at 180 days by Q-PCR and flow-cytometry in all but one case (Figure). 7 patients progressed after day 100, which was accompanied either by loss of engineered T cells or loss of tumor antigen. Detailed phenotyping and functional analysis of engineered T cells, and correlates with clinical responses, is underway. Summary This is the first clinical evaluation of engineered T cells in the MM setting. Infusions are safe, well tolerated, and are associated with encouraging responses in a high risk myeloma population. A study evaluating the engineered T cells in a non-transplant study is underway. Disclosures: Stadtmauer: Celgene: Consultancy. Binder-Scholl:Adaptimmune: Employment. Smethurst:Adaptimmune: Employment. Brewer:Adaptimmune: Employment. Bennett:Adaptimmune: Employment. Gerry:Adaptimmune: Employment. Pumphrey:Adaptimmune: Employment. Tayton-Martin:Adaptimmune: Employment. Ribeiro:Adaptimmune: Employment. Levine:Novartis: cell and gene therapy IP, cell and gene therapy IP Patents & Royalties. Jakobsen:Adaptimmune: Employment. Kalos:Novartis corporation: CART19 technology, CART19 technology Patents & Royalties; Adaptive biotechnologies: Member scientific advisory board , Member scientific advisory board Other. June:Novartis: Patents & Royalties, Research Funding.


JCI Insight ◽  
2018 ◽  
Vol 3 (8) ◽  
Author(s):  
Alfred L. Garfall ◽  
Edward A. Stadtmauer ◽  
Wei-Ting Hwang ◽  
Simon F. Lacey ◽  
Jan Joseph Melenhorst ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 797-797 ◽  
Author(s):  
Edward A. Stadtmauer ◽  
Dan T. Vogl ◽  
Nicole A. Aqui ◽  
Aaron P. Rapoport ◽  
Kenyetta Macdonald ◽  
...  

Abstract Abstract 797 Introduction: Epidemics of influenza A virus strains have been associated with a mortality rate of 0.1% and hospitalization of approximately 200,000 people per year in the United States. The recent pandemic of swine-origin H1N1 has further emphasized the need to protect the population. Patients with multiple myeloma (MM) are particularly vulnerable to flu infection from altered humoral and cellular immunity from the disease as well as from the immunosuppressive chemotherapies, corticosteroids and radiation frequently used for treatment. In particular, high dose melphalan and ASCT, a common and efficacious treatment as part of first-line or salvage therapy for MM, is associated with post-ASCT immune suppression by virtue of low T-cell numbers, poor T-cell function and immune paresis. Despite anti-infection vaccination after ASCT, pneumococcal pneumonia and influenza are common. In an attempt to improve this outcome we previously showed that adoptive transfer of in-vivo PCV vaccine-primed and ex-vivo (anti-CD3/anti-CD28) co-stimulated autologous T cells (aT-cells) early (within 14 days) post-ASCT increased CD4 and CD8 T cell counts and induced pneumococcal conjugate vaccine-directed T and B-cell responses [Rapoport et al, Nature Medicine, 2005]. Patients who did not receive pre-collection vaccination did not show this response despite infusion of co-stimulated T-cells and post-ASCT vaccination. In the current clinical trial we investigated whether pre-harvest flu vaccination would result in a similar improvement in immunity. Methods: We performed a randomized open-label single center study of two influenza vaccine schedules for patients with high-risk MM responding after conventional induction therapy. Of 21 patients enrolled, 11 were randomly assigned to Group X and received pre- and d+14 post-transplant flu vaccine with the commercially available influenza vaccine (Fluzone) produced by Aventis Pasteur. The 10 patients in Group Y received only post-transplant vaccination. All patients received T-cell harvest, high dose melphalan and ASCT and aT-cell infusion. Immunological assessment (T-cell and B-cell subsets, T-cell repertoire, T-ELISPOT, CFSE-CBC, antibody HAI, B-ELISPOT) was conducted d+60, 100 and 180. The primary study endpoint is the immune response to influenza as measured by the serotype-specific influenza antibody response on day 100 using a standard hemagglutination inhibition assay (HAI) optimized for the vaccine administered each year. Results: The median age was 55 (range 37-68), 13 M, 8 F, 18 Caucasian, 2 African American, 1 Hispanic, IgG 57%, IgA 33%, light chain 10%. With a median follow-up of 1 year, 86% (18/21) patients are alive with 48% (10/21) alive in remission. No difference in these parameters was seen between both Groups. HAI titers, however were higher at all three time points (d+60, 100, 180) for Group X when compared to Group Y both for H3N2 (p= 0.04) and for H1N1 (p=0.07). HAI titers for Group Y remained near baseline throughout all time points while Group X peaked day 60 and remained elevated day 180. Analysis of other immune assessment is ongoing and will be presented. Conclusion: Preliminary analysis of this randomized trial of flu vaccine schedule for MM patients undergoing ASCT suggests superior flu immune reconstitution when vaccine is administered to prime autologous T-cells prior ASCT and aT-cell infusion. This result adds to our observation that an approach of vaccine priming and co-stimulated autologous T-cell infusion after high dose melphalan and ASCT for MM augments an otherwise poor response in this patient population to pneumococcal and influenza vaccination. Investigation of this approach as a platform for anti-myeloma immune therapy is ongoing. Disclosures: June: University of Pennsylvania: Patents & Royalties.


Sign in / Sign up

Export Citation Format

Share Document