Analysis Of Immune Cell Populations Before and After Autologous Hematopoietic Stem Cell Transplant For Multiple Myeloma: Association With Early Recovery Of Absolute Lymphocyte Count and Progression-Free Survival

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3348-3348
Author(s):  
Theresa Hahn ◽  
Yali Zhang ◽  
Bruno Paiva ◽  
George L. Chen ◽  
Paul K. Wallace ◽  
...  

Abstract The treatment of transplant-eligible, multiple myeloma (MM) patients usually consists of induction therapy followed by autologous hematopoietic stem cell transplant (ASCT). In addition to typical cytogenetic and biochemical prognostic factors, there are other risk factors that predict for relapse and progression after ASCT. This includes absolute lymphocyte count (ALC) recovery (>0.5 cells/μL) by day +15 post ASCT, which has previously been shown to correlate with prolonged progression-free (PFS) and overall survival (OS). In a cohort of 70 MM patients who received high dose melphalan with ASCT between 8/2007 and 9/2012, we performed a comprehensive immunophenotyping panel including 35 T-, B-, NK- and Dendritic Cell (DC)-cell subsets a median of 25 days before ASCT (N=70), at day +30 post-ASCT (N=40) and at day +100 (N=51) post-ASCT. Specifically, the immunophenotyping panel included total numbers of T cells (CD3+, CD4+, CD8+), B cells (CD19+, CD20+), NK cells (CD56+CD16+) and myeloid (CD11c+) and plasmacytoid (CD123+) dendritic cells. T cell subsets included the relative proportions of double positive (CD3+CD4+CD8+), double negative (CD3+CD4-CD8-), CD4+ and CD8+ Naïve (CD3+ CD45RA+ CD45RO- CD27+), CD4+ and CD8+ central memory (CD3+ CD45RA- CD45RO+ CD27+), CD4+ and CD8+ effector memory (CD3+ CD45RA- CD45RO+ CD27-), recent thymic emigrants (CD3+ CD4+ CD45RA+ CD31+), and T-regulatory cells (CD3+ CD4+ CD25(br), CD3+CD4+CD25+CD127(d), CD3+CD4+CD25+CD127-HLADr+). B cell subsets included proportions of naïve (CD19+ CD27-), and memory (CD19+ CD27+, pre-switch CD19+ IgD+ CD27+, post-switch CD19+ IgD- CD27) cells. All patients survived to day +100. We examined MM response to ASCT, recovery of ALC by day +15 post-ASCT, clinically significant infections, PFS and OS. Characteristics in 70 patients pre-ASCT were: median (range) age 60 (28-75) yrs; 54% females; 26% KPS ≥90; response to most recent therapy was 13% in CR/sCR, 40% in VGPR, 41% in PR, 6% stable disease. MM responses at day + 100 included: 51% achieved or maintained CR/sCR, 17% VGPR, 10% PR, 24% SD and 6% progressed. The frequency of different immune cell subsets at either pre-ASCT or day +100 post-ASCT were not associated with CR/sCR status at day 100 post-AHSCT. ALC recovery by day +15 post-ASCT was associated with significantly longer median PFS (651 vs 288 days, P=0.001) and OS (804 vs 391 days, P=0.02) than those who did not recover ALC by day 15. For patients showing early recovery of ALC, the absolute numbers of T-cells pre-ASCT (P=0.08), as well as the proportion of CD4+ naïve (P=0.07) and CD8+ NK (CD8+/CD16+/CD56+, P=0.06) populations tended to be higher, whereas the absolute counts of gamma-delta T cells (P=0.02), CD4+ helper (P=0.03, See Figure), CD8+ effector (P=0.02) T-cells and total B-cell count (P=0.05) were significantly higher as compared to patients who did not achieve ALC recovery by day +15. Of note, within the T-cell compartment, CD4+ central memory (P=0.02, see Figure), CD8+ central memory (P=0.05), T-regulatory (T reg) (P=0.02, CD4+25+) populations were significantly lower. The distribution of different immune cell subsets pre-ASCT was not associated with development of clinically significant infections before day +100 post-AHCT; however, the proportion of memory B cells measured at day+100 was significantly higher (p=0.02) in patients who had experienced an infection before day +100. In the present study we show that the distribution of different immune cell populations correlates with ALC recovery in MM. Moreover, we also confirmed the association of ALC recovery with prolonged PFS and OS. The increase in the relative proportions of CD4+ and CD8+ effector cell subpopulations and the decrease in CD4+ and CD8+ central memory and T-reg populations demonstrate that the pre-ASCT effector cell phenotype contributes to faster ALC recovery, and improved PFS/OS. Therapeutic strategies to alter the pre-ASCT and/or day + 100 immune phenotype may improve outcomes after ASCT.Figure 1Figure 1. Figure 2Figure 2. Disclosures: McCarthy: Janssen: Honoraria; Celgene: Consultancy.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4898-4898
Author(s):  
John E Coda ◽  
Kadee Raser ◽  
Sarah M. Anand ◽  
Monalisa Ghosh ◽  
John Maciejewski ◽  
...  

Abstract Background Infections remain a common cause of morbidity and mortality following hematopoietic stem cell transplant (HSCT). Pneumocystis jiroveci pneumonia (PJP) is an opportunistic infection that can occur in HSCT patients, although the association is best demonstrated in allogeneic HSCT, occurring less commonly in autologous HSCT. However, reports on PJP incidence, timing of infection, and outcomes among autologous HSCT cohorts are limited. Furthermore, while current guidelines recommend 3-6 months of prophylaxis against PJP following autologous HSCT, the optimal duration and even necessity of prophylaxis is not well established. Patients and Methods We performed a retrospective analysis of all consecutive patients who had autologous HSCT at the University of Michigan Blood and Marrow Transplantation program over a 20-year period from 1/1/2000 through 12/31/2019. The cohort consisted of a total of 2082 patients, 1221 male (58.6%), with median age 56 (range 10 months - 77 years, 91.2% ≥ 18 years). Records were searched for use of PJP prophylaxis over 6-month and 2-year follow-up periods post-HCT to determine rates of prophylaxis and choice of agent. Cases of polymerase-chain reaction (PCR)-confirmed PJP occurring within two years of HSCT were identified. The timing, clinical and laboratory features at diagnosis, use of concurrent immunosuppression, treatment, and outcomes were determined. Results Of the 2082 patients undergoing autologous HCT, 704 patients (33.8%) received PJP prophylaxis in the first 6 months following transplant. Prophylaxis rates varied over time, ranging from 14.6% to 80.0% when calculated by year of transplant (Figure 1). Trimethoprim-sulfamethoxazole (TMP-SMX) was the most used prophylaxis agent (70.3%), followed by inhaled pentamidine (31.8%), with intravenous pentamidine (8.1%), dapsone (7.1%), and atovaquone (2.6%) being used less frequently. There were 9 cases of PJP identified in our cohort, with an incidence of 0.43%. There were 6 males, with median age of 50 (range 34 - 69). Cases occurred a median of 126 days following HSCT (range 65 - 496), with 4 cases occurring after 6 months. None of the patients were on PJP prophylaxis at the time of diagnosis, and only 2 patients had received prophylaxis at any point after transplant. In 8 of 9 cases, patients were receiving concurrent pharmacologic immunosuppression in the form of steroids or maintenance brentuximab (Table 1). All patients presented with symptoms compatible with PJP, most often with fevers, dyspnea, and cough. Diagnosis was made by PCR from bronchoalveolar lavage specimen in 8 cases, and from sputum sample in 1 case. All patients were lymphopenic at the time of diagnosis, with median absolute lymphocyte count of 400 cells/µL (range 200 - 1100). Patients were most often treated with TMP-SMX. Three patients required transfer to the intensive care unit and 2 were intubated. Ultimately, 2 patients died from PJP infection; the remaining 7 recovered (Table 2). Conclusion Our analysis reveals that among a large cohort, incidence of PJP following autologous HSCT is low. This was the case even with relatively modest rates of PJP prophylaxis in the first 6 months following transplant. Most cases of PJP occurred in patients receiving additional immunosuppression and often occurred late following transplant. Figure 1 Figure 1. Disclosures Pianko: Karyopharm: Honoraria.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2913-2913
Author(s):  
Ashley Rose ◽  
Quinto J Gesiotto ◽  
Leidy Isenalumhe ◽  
Farhad Khimani ◽  
Hien D. Liu ◽  
...  

Abstract Introduction: The standard of care for relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) is salvage therapy followed by autologous hematopoietic stem cell transplant (auto-HCT). Pre-apheresis absolute lymphocyte count (PA-ALC) is an independent prognostic factor for overall survival (OS) after transplant. We aimed to evaluate the effect of absolute lymphocyte count following auto-HCT and hypothesized that a higher post-transplant ALC at day +15 (PT-ALC) correlates with improved OS. Methods: A retrospective review was performed on patients with R/R cHL who underwent auto-HCT at Moffitt Cancer Center from 2000-2020. The following patient characteristics were collected: age at diagnosis, gender, initial stage and presence of B symptoms. Pretransplant variables including chemotherapy, number of cycles, response to therapy, and time from last chemo to apheresis were collected. Receiver-operator characteristics (ROC) curve was used to identify the ideal PT-ALC to predict overall survival. Patients were then identified as high ALC versus low ALC. Mann-Whitney, Pearson Chi-square, and Fisher exact test were used to compare baseline characteristics between the two groups. Univariate analysis of overall survival was done using Log-rank testing and Kaplan-Meier curve. Cox-regression analysis was used to evaluate the factors affecting OS. Results: A total of 259 patients were included in the study, with a median age of 35 years (range 14-76). ROC curve was used to identify the ideal PT-ALC affecting OS, and a cutoff value of 300/uL was determined (AUC 0.60; 95% CI: 0.52-0.68, Figure 1). In this cohort, 52 patients (16.6%) had low PT-ALC and 207 patients (65.9%) had high PT-ALC. There was no significant difference between the two groups in regards to patient age, gender, histology type, stage at presentation, number of salvage cycles, number of CD34 cells collected, or number of days required for apheresis. Patients with a high PT-ALC had higher pre-apheresis ALC (p<0.001). There was a trend toward significance with patients with high PT-ALC receiving non-chemotherapy salvage regimens (p=0.07, Table 1). However, PA-ALC was significantly higher in non-chemotherapy regimen (p=0.007). Patients with high PT-ALC had a longer OS after transplant than those with low PT-ALC, with median OS 11.8 years and 7.7 years, respectively (p=0.012, Figure 2). On multivariate analysis, the only factor associated with improved OS was high PT-ALC (p=0.015, Table 2). Conclusions: High PA-ALC and high PT-ALC are both independent prognostic factors for longer OS in patients with relapsed/refractory Hodgkin lymphoma after auto-HCT. High PA-ALC lead to higher PT-ALC. Although most of our patients received chemotherapy as salvage therapy prior to transplant, there was a trend toward higher PT-ALC in patients who received non-chemotherapy regimens. Future studies are required to determine the role of non-chemotherapy salvage regimens in improving lymphocyte counts during the peri-transplant period and, hence, improved survival. Figure 1 Figure 1. Disclosures Gaballa: Beigene: Consultancy; TG therapeutics: Consultancy, Speakers Bureau; Epizyme: Consultancy, Research Funding; Adaptive Biotechnologies: Research Funding; ADC Therapeutics: Consultancy. Chavez: BMS: Speakers Bureau; Merk: Research Funding; ADC Therapeutics: Consultancy, Research Funding; MorphoSys, Bayer, Karyopharm, Kite, a Gilead Company, Novartis, Janssen, AbbVie, TeneoBio, and Pfizer: Consultancy; MorphoSys, AstraZeneca, BeiGene, Genentech, Kite, a Gilead Company, and Epizyme: Speakers Bureau; AstraZeneca: Research Funding. Shah: Bristol-Myers Squibb/Celgene: Consultancy, Other: Expenses; Pharmacyclics/Janssen: Honoraria, Other: Expenses; Acrotech/Spectrum: Honoraria; Incyte: Research Funding; BeiGene: Consultancy, Honoraria; Jazz Pharmaceuticals: Research Funding; Kite, a Gilead Company: Consultancy, Honoraria, Other: Expenses, Research Funding; Precision Biosciences: Consultancy; Amgen: Consultancy; Pfizer: Consultancy, Other: Expenses; Novartis: Consultancy, Other: Expenses; Servier Genetics: Other; Adaptive Biotechnologies: Consultancy. Pinilla Ibarz: Sellas: Other: ), patents/royalties/other intellectual property; MEI, Sunesis: Research Funding; AbbVie, Janssen, AstraZeneca, Takeda: Speakers Bureau; AbbVie, Janssen, AstraZeneca, Novartis, TG Therapeutics, Takeda: Consultancy, Other: Advisory. Sokol: Kyowa-Kirin: Membership on an entity's Board of Directors or advisory committees; Dren Bio: Membership on an entity's Board of Directors or advisory committees. Saeed: Kite Pharma: Consultancy, Other: investigator; sano-aventis U.S.: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb Company: Consultancy; Nektar Therapeutics: Consultancy, Other: research investigator; Other-TG therapeutics: Consultancy, Other: investigator; Other-Epizyme, Inc.: Consultancy; Janssen Pharmaceutica Products, LP: Consultancy, Other: investigator; Celgene Corporation: Consultancy, Other: investigator; MEI Pharma Inc: Consultancy, Other: investigator; MorphoSys AG: Consultancy, Membership on an entity's Board of Directors or advisory committees; Other-Secura Bio, Inc.: Consultancy; Seattle Genetics, Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1704-1704
Author(s):  
Ribhu Nayar ◽  
Mollie M Jurewicz ◽  
Sonal Jangalwe ◽  
Hannah Bader ◽  
Kimberly M Cirelli ◽  
...  

Abstract Background Approximately 50% of AML patients relapse following allogeneic hematopoietic stem cell transplant therapy, leaving them with very few treatment options (Rautenberg et al. (2019) Int. J. Mol. Sci. 20:228). Rare patients who naturally develop a minor antigen-specific graft-versus-leukemia T cell response show substantially lower relapse rates (Marijt et al. (2003) Proc. Natl. Acad. Sci. U.S.A. 100:2742-2747; Spierings et al. (2013) Biol. Blood Marrow Transplant. 19:1244-1253). HA-2 (YIGEVLVSV, genotype RS_61739531 C/C or T/C) is an HLA-A*02:01- and haematopoietically-restricted minor histocompatibility antigen derived from the class I myosin protein, MYO1G (Pierce et al. (2001) J. Immunol. 167:3223-3230). Patients receiving donor lymphocyte infusion from HA-2-mismatched donors who develop HA-2-specific T cells show a graft vs leukemia response and often experience long-term remission (Marijt et al. (2003) Proc. Natl. Acad. Sci. U.S.A. 100:2742-2747), making HA-2 an ideal candidate for TCR-engineered T cell immunotherapy of liquid tumors. Methods Using TScan's proprietary ReceptorScan platform, we discovered 1,302 HA-2-specific TCRs by screening 237 million naïve CD8 + T cells from 5 healthy HA-2-negative donors. We evaluated these TCRs using our proprietary DexScan platform to select the 15 TCRs with the highest surface expression and greatest affinity for the HA-2 peptide when transferred into primary human T cells. We further tested each TCR individually in our clinical vector backbone for surface expression, selective cytotoxicity, cytokine production, and proliferation using a panel of cell lines that express varying levels of HLA-A*02:01 and MYO1G. Finally, the top 5 TCRs were evaluated for alloreactivity using an array-based screen assessing 108 MHC-I molecules individually, and for off-target cross-reactivity using our proprietary genome-wide TargetScan platform. A lead TCR with limited alloreactivity and a narrow off-target profile was selected as our lead TSC-101 TCR. The avidity of TSC-101 for its putative off-targets was further measured in peptide-pulsed experiments to better appreciate the toxicity risks associated with our lead clinical candidate. Results and Conclusion Of the 1,302 HA-2-specific TCRs identified by our ReceptorScan platform, we identified TSC-101 as the most active TCR. TSC-101 displayed no alloreactivity to 107/108 HLAs tested and limited off-target risks in a genome-wide screens. Potential off-target peptides identified for TSC-101 displayed extremely weak avidities, predicting an absence of toxicity risks for our clinical candidate. Based on these results, TSC-101 has been advanced to IND-enabling activities to prepare for first-in-human testing in 2022. To our knowledge, this is the first clinical grade HA-2-specifc TCR being developed for immunotherapy for liquid tumors. Disclosures Macbeath: TScan Therapeutics: Current Employment, Current equity holder in publicly-traded company.


2021 ◽  
Vol 9 (7) ◽  
pp. e002968
Author(s):  
Shelley Herbrich ◽  
Natalia Baran ◽  
Tianyu Cai ◽  
Connie Weng ◽  
Marisa J L Aitken ◽  
...  

BackgroundAcute myeloid leukemia (AML) stem cells (LSCs) are capable of surviving current standard chemotherapy and are the likely source of deadly, relapsed disease. While stem cell transplant serves as proof-of-principle that AML LSCs can be eliminated by the immune system, the translation of existing immunotherapies to AML has been met with limited success. Consequently, understanding and exploiting the unique immune-evasive mechanisms of AML LSCs is critical.MethodsAnalysis of stem cell datasets and primary patient samples revealed CD200 as a putative stem cell–specific immune checkpoint overexpressed in AML LSCs. Isogenic cell line models of CD200 expression were employed to characterize the interaction of CD200+ AML with various immune cell subsets both in vitro and in peripheral blood mononuclear cell (PBMC)–humanized mouse models. CyTOF and RNA-sequencing were performed on humanized mice to identify novel mechanisms of CD200-mediated immunosuppression. To clinically translate these findings, we developed a fully humanized CD200 antibody (IgG1) that removed the immunosuppressive signal by blocking interaction with the CD200 receptor while also inducing a potent Fc-mediated response. Therapeutic efficacy of the CD200 antibody was evaluated using both humanized mice and patient-derived xenograft models.ResultsOur results demonstrate that CD200 is selectively overexpressed in AML LSCs and is broadly immunosuppressive by impairing cytokine secretion in both innate and adaptive immune cell subsets. In a PBMC-humanized mouse model, CD200+ leukemia progressed rapidly, escaping elimination by T cells, compared with CD200− AML. T cells from mice with CD200+ AML were characterized by an abundance of metabolically quiescent CD8+ central and effector memory cells. Mechanistically, CD200 expression on AML cells significantly impaired OXPHOS metabolic activity in T cells from healthy donors. Importantly, CD200 antibody therapy could eliminate disease in the presence of graft-versus-leukemia in immune competent mice and could significantly improve the efficacy of low-intensity azacitidine/venetoclax chemotherapy in immunodeficient hosts.ConclusionsOverexpression of CD200 is a stem cell–specific marker that contributes to immunosuppression in AML by impairing effector cell metabolism and function. CD200 antibody therapy is capable of simultaneously reducing CD200-mediated suppression while also engaging macrophage activity. This study lays the groundwork for CD200-targeted therapeutic strategies to eliminate LSCs and prevent AML relapse.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1068-1068 ◽  
Author(s):  
Thomas J. Manley ◽  
Tori Yamamoto ◽  
Lisa Luy ◽  
Thomas Jones ◽  
Michael Boeckh ◽  
...  

Abstract Human cytomegalovirus (CMV) may cause severe disease in immunosuppressed hosts including hematopoietic stem cell transplant recipients. The components of host immunity that provide protection from progressive infection have not been completely defined. Studies in immunocompromised CMV-infected humans, and in mice infected with murine cytomegalovirus (mCMV), have shown that recovery of class I major histocompatibility complex (MHC) restricted CD8+ cytotoxic T lymphocytes (CTL) that recognize viral peptides play a key role in containing viral replication. The importance of CTL specific for individual antigens in immune control of CMV has been difficult to determine because cells permissively infected with human CMV express over 160 viral proteins during the (IE), early (E), and late (L) phases of replication. Recent studies in immunocompetent humans have identified significant frequencies of CTL specific for multiple CMV antigens expressed at all stages of viral replication, despite the presence of viral proteins that interfere with class I antigen presentation. Thus, analysis of CMV-specific T cell immunity using one or a few peptides is likely to underestimate the magnitude and diversity of the host response. We have developed a genetic approach for characterizing CMV antigens that concurrently identifies the HLA restricting allele and enables rapid determination of the minimal epitope derived from any CMV gene. To evaluate this approach, PBMC from seropositive individuals were stimulated once in vitro with autologous fibroblasts infected with the RV798 strain of CMV, which lacks all class I immune evasion genes and enables display of all potentially antigenic peptides. The T cells were then screened against a panel of COS cells transfected with a plasmid library containing a majority of CMV ORFs and with each of the HLA alleles of the donor. Twenty-two CMV genes that were predominantly expressed at IE or E stages of infection were identified to encode antigens recognized by CTL from 4 normal donors. The median number of antigens recognized in each donor was 8 (range 4–12). Seventeen CMV peptides presented by a variety of common HLA class I molecules including HLA-C were subsequently mapped and two epitopes were found to be derived from alternative translation products or processing mechanisms. Memory T cells from other CMV seropositive individuals that shared the HLA restricting allele also recognized these novel epitopes. This genome scan was used successfully to identify the repertoire of CMV antigens recognized by CMV-specific CTL generated from CMV seropositive hematopoietic stem cell transplant donors and to determine which responses were transferred to the respective recipient post transplant. CTL specific for a broad repertoire of viral antigens comparable to that in the donor were found in some transplant recipients, while in others the response was dramatically restricted compared to the donor. These results further define the broad specificity of the CMV-specific CTL response in seropositive donors, enable comprehensive monitoring of the recovery of CMV-specific CTL in immunocompromised patients at risk for CMV disease, and may be useful for defining the specificity of CTL responses that correlate with control of virus replication.


Sign in / Sign up

Export Citation Format

Share Document