87 results on '"Mark B. Geyer"'
Search Results
2. Depletion of high-content CD14+ cells from apheresis products is critical for successful transduction and expansion of CAR T cells during large-scale cGMP manufacturing
- Author
-
Xiuyan Wang, Oriana Borquez-Ojeda, Jolanta Stefanski, Fang Du, Jinrong Qu, Jagrutiben Chaudhari, Keyur Thummar, Mingzhu Zhu, Ling-bo Shen, Melanie Hall, Paridhi Gautam, Yongzeng Wang, Brigitte Sénéchal, Devanjan Sikder, Prasad S. Adusumilli, Renier J. Brentjens, Kevin Curran, Mark B. Geyer, Sham Mailankhody, Roisin O’Cearbhaill, Jae H. Park, Craig Sauter, Susan Slovin, Eric L. Smith, and Isabelle Rivière
- Subjects
CAR T cell ,cGMP ,clinical grade ,large-scale manufacturing ,monocyte depletion ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin’s lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success.
- Published
- 2021
- Full Text
- View/download PDF
3. Pediatric-inspired chemotherapy incorporating pegaspargase is safe and results in high rates of minimal residual disease negativity in adults up to age 60 with Philadelphia chromosome-negative acute lymphoblastic leukemia
- Author
-
Mark B. Geyer, Ellen K. Ritchie, Arati V. Rao, Shreya Vemuri, Jessica Flynn, Meier Hsu, Sean M. Devlin, Mikhail Roshal, Qi Gao, Madhulika Shukla, Jose M. Salcedo, Peter Maslak, Martin S. Tallman, Dan Douer, and Jae H. Park
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Administration of pediatric-inspired chemotherapy to adults up to age 60 with acute lymphoblastic leukemia (ALL) is challenging in part due to toxicities of asparaginase as well as myelosuppression. We conducted a multicenter phase II clinical trial (NCT01920737) investigating a pediatric-inspired regimen, based on the augmented arm of the Children’s Cancer Group 1882 protocol, incorporating 6 doses of pegaspargase 2000 IU/m2, rationally synchronized to avoid overlapping toxicity with other agents. We treated 39 adults ages 20-60 years (median, 38 years) with newly-diagnosed ALL (n=31) or lymphoblastic lymphoma (n=8). Grade 3-4 hyperbilirubinemia occurred frequently and at higher rates in patients 40-60 (n=18) vs 18-39 (n=21) years (44 vs 10%, p=0.025). However, 8/9 patients re-challenged with pegaspargase did not experience recurrent grade 3-4 hyperbilirubinemia. Grade 3-4 hypertriglyceridemia and hypofibrinogenemia were common (each 59%). Asparaginase activity at 7-days post-infusion reflected levels associated with adequate asparagine depletion, even among those with antibodies to pegaspargase. Complete response (CR)/CR with incomplete hematologic recovery was observed post-induction in 38/39 (97%) patients. Among patients with ALL, rates of MRD negativity by multiparameter flow cytometry were 33% and 83% following Induction Phase I and Phase II, respectively. Event-free and overall survival at 3 years (67.8 and 76.4%) compare favorably to outcomes observed in other series. These results demonstrate pegaspargase can be administered in the context of intensive multi-agent chemotherapy to adults age ≤60 with manageable toxicity. This regimen may serve as an effective backbone into which novel agents may be incorporated in future frontline studies.
- Published
- 2020
- Full Text
- View/download PDF
4. Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia
- Author
-
Wenbin Xiao, Aaron D. Goldberg, Christopher A. Famulare, Sean M. Devlin, Nghia T. Nguyen, Sinnifer Sim, Charlene C. Kabel, Minal A. Patel, Erin M. McGovern, Akshar Patel, Jessica Schulman, Andrew J. Dunbar, Zachary D. Epstein-Peterson, Kamal N. Menghrajani, Bartlomiej M. Getta, Sheng F. Cai, Mark B. Geyer, Jacob L. Glass, Justin Taylor, Aaron D. Viny, Ross L. Levine, Yanming Zhang, Sergio A. Giralt, Virginia Klimek, Martin S. Tallman, and Mikhail Roshal
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Measurable residual disease is associated with inferior outcomes in patients with acute myeloid leukemia (AML). Measurable residual disease monitoring enhances risk stratification and may guide therapeutic intervention. The European LeukemiaNet working party recently came to a consensus recommendation incorporating leukemia associated immunophenotype-based different from normal approach by multi-color flow cytometry for measurable residual disease evaluation. However, the analytical approach is highly expertise-dependent and difficult to standardize. Here we demonstrate that loss of plasmacytoid dendritic cell differentiation after 7+3 induction in AML is highly specific for measurable residual disease positivity (specificity 97.4%) in a uniformly treated patient cohort. Moreover, loss of plasmacytoid dendritic cell differentiation as determined by a blast-to-plasmacytoid dendritic cell ratio >10 was strongly associated with inferior overall and relapse-free survival (RFS) [Hazard ratio 2.79, 95% confidence interval (95%CI): 0.98-7.97; P=0.077) and 3.83 (95%CI: 1.51-9.74; P=0.007), respectively), which is similar in magnitude to measurable residual disease positivity. Importantly, measurable residual disease positive patients who reconstituted plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio
- Published
- 2019
- Full Text
- View/download PDF
5. Inotuzumab ozogamicin as chemotherapy-sparing salvage in a 67-year-old man with primary refractory B-cell acute lymphoblastic leukemia with high-risk genomic features
- Author
-
Xiaochuan Yang, Filiz Sen, and Mark B. Geyer
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Older adults with acute lymphoblastic leukemia (ALL) continue to have a poor prognosis, in part due to greater chemotherapy-related toxicities. We herein report a 67-year-old man with Philadelphia chromosome (Ph)-negative B-cell ALL, who exhibited refractoriness to 3 different regimens of induction chemotherapy and experienced multiple complications including intracranial bleeding and respiratory failure, who achieved minimal residual disease (MRD)-negative complete response (CR) after a single cycle of inotuzumab ozogamicin (IO). His ALL was characterized by several high-risk mutations, which may have contributed to chemotherapy-refractory disease. Our case supports incorporating IO into front-line induction regimens for older adults with high-risk B-cell ALL. Keywords: Acute lymphoblastic leukemia, Inotuzumab ozogamicin, Older adults, Immunotherapy
- Published
- 2019
- Full Text
- View/download PDF
6. Molecular predictors of immunophenotypic measurable residual disease clearance in acute myeloid leukemia
- Author
-
Maximilian Stahl, Andriy Derkach, Noushin Farnoud, Jan Philipp Bewersdorf, Troy Robinson, Christopher Famulare, Christina Cho, Sean Devlin, Kamal Menghrajani, Minal A. Patel, Sheng F. Cai, Linde A. Miles, Robert L. Bowman, Mark B. Geyer, Andrew Dunbar, Zachary D. Epstein‐Peterson, Erin McGovern, Jessica Schulman, Jacob L. Glass, Justin Taylor, Aaron D. Viny, Eytan M. Stein, Bartlomiej Getta, Maria E. Arcila, Qi Gao, Juliet Barker, Brian C. Shaffer, Esperanza B. Papadopoulos, Boglarka Gyurkocza, Miguel‐Angel Perales, Omar Abdel‐Wahab, Ross L. Levine, Sergio A. Giralt, Yanming Zhang, Wenbin Xiao, Nidhi Pai, Elli Papaemmanuil, Martin S. Tallman, Mikhail Roshal, and Aaron D. Goldberg
- Subjects
Hematology - Abstract
Measurable residual disease (MRD) is a powerful prognostic factor in acute myeloid leukemia (AML). However, pre-treatment molecular predictors of immunophenotypic MRD clearance remain unclear. We analyzed a dataset of 211 patients with pre-treatment next-generation sequencing who received induction chemotherapy and had MRD assessed by serial immunophenotypic monitoring after induction, subsequent therapy, and allogeneic stem cell transplant (allo-SCT). Induction chemotherapy led to MRD- remission, MRD+ remission, and persistent disease in 35%, 27%, and 38% of patients, respectively. With subsequent therapy, 34% of patients with MRD+ and 26% of patients with persistent disease converted to MRD-. Mutations in CEBPA, NRAS, KRAS, and NPM1 predicted high rates of MRD- remission, while mutations in TP53, SF3B1, ASXL1, and RUNX1 and karyotypic abnormalities including inv (3), monosomy 5 or 7 predicted low rates of MRD- remission. Patients with fewer individual clones were more likely to achieve MRD- remission. Among 132 patients who underwent allo-SCT, outcomes were favorable whether patients achieved early MRD- after induction or later MRD- after subsequent therapy prior to allo-SCT. As MRD conversion with chemotherapy prior to allo-SCT is rarely achieved in patients with specific baseline mutational patterns and high clone numbers, upfront inclusion of these patients into clinical trials should be considered.
- Published
- 2022
- Full Text
- View/download PDF
7. Asciminib monotherapy in patients with CML-CP without BCR::ABL1 T315I mutations treated with at least two prior TKIs: 4-year phase 1 safety and efficacy results
- Author
-
Michael J. Mauro, Timothy P. Hughes, Dong-Wook Kim, Delphine Rea, Jorge E. Cortes, Andreas Hochhaus, Koji Sasaki, Massimo Breccia, Moshe Talpaz, Oliver Ottmann, Hironobu Minami, Yeow Tee Goh, Daniel J. DeAngelo, Michael C. Heinrich, Valle Gómez-García de Soria, Philipp le Coutre, Francois-Xavier Mahon, Jeroen J. W. M. Janssen, Michael Deininger, Naranie Shanmuganathan, Mark B. Geyer, Silvia Cacciatore, Fotis Polydoros, Nithya Agrawal, Matthias Hoch, Fabian Lang, Hematology, CCA - Cancer biology and immunology, and CCA - Imaging and biomarkers
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
Asciminib is approved for patients with Philadelphia chromosome–positive chronic-phase chronic myeloid leukemia (CML-CP) who received ≥2 prior tyrosine kinase inhibitors or have the T315I mutation. We report updated results of a phase 1, open-label, nonrandomized trial (NCT02081378) assessing the safety, tolerability, and antileukemic activity of asciminib monotherapy 10–200 mg once or twice daily in 115 patients with CML-CP without T315I (data cutoff: January 6, 2021). After ≈4-year median exposure, 69.6% of patients remained on asciminib. The most common grade ≥3 adverse events (AEs) included increased pancreatic enzymes (22.6%), thrombocytopenia (13.9%), hypertension (13.0%), and neutropenia (12.2%); all-grade AEs (mostly grade 1/2) included musculoskeletal pain (59.1%), upper respiratory tract infection (41.7%), and fatigue (40.9%). Clinical pancreatitis and arterial occlusive events (AOEs) occurred in 7.0% and 8.7%, respectively. Most AEs occurred during year 1; the subsequent likelihood of new events, including AOEs, was low. By data cutoff, among patients without the indicated response at baseline, 61.3% achieved BCR::ABL1 ≤ 1%, 61.6% achieved ≤0.1% (major molecular response [MMR]), and 33.7% achieved ≤0.01% on the International Scale. MMR was maintained in 48/53 patients who achieved it and 19/20 who were in MMR at screening, supporting the long-term safety and efficacy of asciminib in this population.
- Published
- 2023
- Full Text
- View/download PDF
8. Data from BCMA-Targeted CAR T-cell Therapy plus Radiotherapy for the Treatment of Refractory Myeloma Reveals Potential Synergy
- Author
-
Renier J. Brentjens, Ola Landgren, Isabelle Riviere, Andreas Rimner, Jonathan Landa, Bianca D. Santomasso, Elena Mead, Aaron D. Goldberg, Mark B. Geyer, Anthony F. Daniyan, Terence J. Purdon, Brigitte Senechal, Xiuyan Wang, Mette Staehr, Sham Mailankody, and Eric L. Smith
- Abstract
We present a case of a patient with multiply relapsed, refractory myeloma whose clinical course showed evidence of a synergistic abscopal-like response to chimeric antigen receptor (CAR) T-cell therapy and localized radiotherapy (XRT). Shortly after receiving B-cell maturation antigen (BCMA)–targeted CAR T-cell therapy, the patient required urgent high-dose steroids and XRT for spinal cord compression. Despite the steroids, the patient had a durable systemic response that could not be attributed to XRT alone. Post-XRT findings included a second wave of fever and increased CRP and IL6, beginning 21 days after CAR T cells, which is late for cytokine-release syndrome from CAR T-cell therapy alone on this trial. Given this response, which resembled cytokine-release syndrome, immediately following XRT, we investigated changes in the patient's T-cell receptor (TCR) repertoire over 10 serial time points. Comparing T-cell diversity via Morisita's overlap indices (CD), we discovered that, although the diversity was initially stable after CAR T-cell therapy compared with baseline (CD = 0.89–0.97, baseline vs. 4 time points after CAR T cells), T-cell diversity changed after the conclusion of XRT, with >30% newly expanded TCRs (CD = 0.56–0.69, baseline vs. 4 time points after XRT). These findings suggest potential synergy between radiation and CAR T-cell therapies resulting in an abscopal-like response.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Data from BCMA-Targeted CAR T-cell Therapy plus Radiotherapy for the Treatment of Refractory Myeloma Reveals Potential Synergy
- Author
-
Renier J. Brentjens, Ola Landgren, Isabelle Riviere, Andreas Rimner, Jonathan Landa, Bianca D. Santomasso, Elena Mead, Aaron D. Goldberg, Mark B. Geyer, Anthony F. Daniyan, Terence J. Purdon, Brigitte Senechal, Xiuyan Wang, Mette Staehr, Sham Mailankody, and Eric L. Smith
- Abstract
Supplementary Data Combined
- Published
- 2023
- Full Text
- View/download PDF
10. Biomarkers and cardiovascular outcomes in chimeric antigen receptor T-cell therapy recipients
- Author
-
Syed S Mahmood, Peter A Riedell, Stephanie Feldman, Gina George, Stephen A Sansoterra, Thomas Althaus, Mahin Rehman, Elena Mead, Jennifer E Liu, Richard B Devereux, Jonathan W Weinsaft, Jiwon Kim, Lauren Balkan, Tarek Barbar, Katherine Lee Chuy, Bhisham Harchandani, Miguel-Angel Perales, Mark B Geyer, Jae H Park, M Lia Palomba, Roni Shouval, Ana A Tomas, Gunjan L Shah, Eric H Yang, Daria L Gaut, Michael V Rothberg, Evelyn M Horn, John P Leonard, Koen Van Besien, Matthew J Frigault, Zhengming Chen, Bhoomi Mehrotra, Tomas G Neilan, and Richard M Steingart
- Subjects
Cardiology and Cardiovascular Medicine - Abstract
Aims Chimeric antigen receptor T-cell therapy (CAR-T) harnesses a patient’s immune system to target cancer. There are sparse existing data characterizing death outcomes after CAR-T-related cardiotoxicity. This study examines the association between CAR-T-related severe cardiovascular events (SCE) and mortality. Methods and results From a multi-centre registry of 202 patients receiving anti-CD19 CAR-T, covariates including standard baseline cardiovascular and cancer parameters and biomarkers were collected. Severe cardiovascular events were defined as a composite of heart failure, cardiogenic shock, or myocardial infarction. Thirty-three patients experienced SCE, and 108 patients died during a median follow-up of 297 (interquartile range 104–647) days. Those that did and did not die after CAR-T were similar in age, sex, and prior anthracycline use. Those who died had higher peak interleukin (IL)-6 and ferritin levels after CAR-T infusion, and those who experienced SCE had higher peak IL-6, C-reactive protein (CRP), ferritin, and troponin levels. The day-100 and 1-year Kaplan–Meier overall mortality estimates were 18% and 43%, respectively, while the non-relapse mortality (NRM) cumulative incidence rates were 3.5% and 6.7%, respectively. In a Cox model, SCE occurrence following CAR-T was independently associated with increased overall mortality risk [hazard ratio (HR) 2.8, 95% confidence interval (CI) 1.6–4.7] after adjusting for age, cancer type and burden, anthracycline use, cytokine release syndrome grade ≥ 2, pre-existing heart failure, hypertension, and African American ancestry; SCEs were independently associated with increased NRM (HR 3.5, 95% CI 1.4–8.8) after adjusting for cancer burden. Conclusion Chimeric antigen receptor T-cell therapy recipients who experience SCE have higher overall mortality and NRM and higher peak levels of IL-6, CRP, ferritin, and troponin.
- Published
- 2023
- Full Text
- View/download PDF
11. Interaction between myelodysplasia-related gene mutations and ontogeny in acute myeloid leukemia: an appraisal of the new WHO and IC classifications and ELN risk stratification
- Author
-
Joseph GW. McCarter, David Nemirovsky, Christopher A. Famulare, Noushin Farnoud, Abhinita S. Mohanty, Zoe S. Stone-Molloy, Jordan Chervin, Brian J. Ball, Zachary D. Epstein-Peterson, Maria E. Arcila, Aaron J. Stonestrom, Andrew Dunbar, Sheng F. Cai, Jacob L. Glass, Mark B. Geyer, Raajit K. Rampal, Ellin Berman, Omar I. Abdel-Wahab, Eytan M. Stein, Martin S. Tallman, Ross L. Levine, Aaron D. Goldberg, Elli Papaemmanuil, Yanming Zhang, Mikhail Roshal, Andriy Derkach, and Wenbin Xiao
- Abstract
Accurate classification and risk stratification is critical for clinical decision making in AML patients. In the newly proposed World Health Organization (WHO) and International Consensus classifications (ICC) of hematolymphoid neoplasms, the presence of myelodysplasia-related (MR) gene mutations is included as one of the diagnostic criteria of AML, myelodysplasia-related (AML-MR), largely based on the assumption that these mutations are specific for AML with an antecedent myelodysplastic syndrome. ICC also prioritizes MR gene mutations over ontogeny (as defined by clinical history). Furthermore, European LeukemiaNet (ELN) 2022 stratifies these MR gene mutations to the adverse-risk group. By thoroughly annotating a cohort of 344 newly diagnosed AML patients treated at Memorial Sloan Kettering Cancer Center (MSKCC), we show that ontogeny assignment based on database registry lacks accuracy. MR gene mutations are frequently seen inde novoAML. Among MR gene mutations, onlyEZH2andSF3B1were associated with an inferior outcome in a univariate analysis. In a multivariate analysis, AML ontogeny had independent prognostic values even after adjusting for age, treatment, allo-transplant and genomic classes or ELN risks. Ontogeny also stratified the outcome of AML with MR gene mutations. Finally,de novoAML with MR gene mutations did not show an adverse outcome. In summary, our study emphasizes the importance of accurate ontogeny designation in clinical studies, demonstrates the independent prognostic value of AML ontogeny and questions the current classification and risk stratification of AML with MR gene mutations.Key pointsBoth ontogeny and genomics show independent prognostic values in AML.The newly proposed myelodysplasia-related gene mutations are neither specific to AML-MRCWHO2016nor predictive for adverse outcomes.Ontogeny stratifies the outcome of AML with myelodysplasia-related gene mutations.
- Published
- 2022
- Full Text
- View/download PDF
12. Depletion of high-content CD14+ cells from apheresis products is critical for successful transduction and expansion of CAR T cells during large-scale cGMP manufacturing
- Author
-
Eric L. Smith, Brigitte Senechal, Craig S. Sauter, Jolanta Stefanski, Fang Du, Prasad S. Adusumilli, Susan F. Slovin, Ling-bo Shen, Devanjan S. Sikder, Jagrutiben Chaudhari, Kevin J. Curran, Renier J. Brentjens, Keyur Thummar, Yongzeng Wang, Melanie Hall, Xiuyan Wang, Isabelle Riviere, Roisin E. O'Cearbhaill, Mark B. Geyer, Sham Mailankhody, Mingzhu Zhu, Jae H. Park, Paridhi Gautam, Jinrong Qu, and Oriana Borquez-Ojeda
- Subjects
CD3 ,Cell ,Phases of clinical research ,monocyte depletion ,Pharmacology ,QH426-470 ,CD19 ,medicine ,clinical grade ,Genetics ,large-scale manufacturing ,Molecular Biology ,Multiple myeloma ,B cell ,biology ,QH573-671 ,business.industry ,Monocyte ,medicine.disease ,Chimeric antigen receptor ,cGMP ,medicine.anatomical_structure ,CAR T cell ,Immunology ,biology.protein ,Molecular Medicine ,business ,Cytology - Abstract
With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin's lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success.
- Published
- 2021
13. Neutropenia in adult acute myeloid leukemia patients represents a powerful risk factor for COVID-19 related mortality
- Author
-
Molly Maloy, Martin S. Tallman, Lindsey E. Roeker, Gunjan L. Shah, Anthony F. Daniyan, Aaron D. Goldberg, Mark B Geyer, Varun Narendra, Andriy Derkach, Justin Jee, Maximilian Stahl, and Anthony R. Mato
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Neutropenia ,Coronavirus disease 2019 (COVID-19) ,medicine.medical_treatment ,medicine.disease_cause ,Article ,03 medical and health sciences ,COVID-19 Testing ,0302 clinical medicine ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Risk factor ,Mechanical ventilation ,SARS-CoV-2 ,business.industry ,COVID-19 ,Cancer ,Adult Acute Myeloid Leukemia ,Hematology ,medicine.disease ,Leukemia, Myeloid, Acute ,Leukemia ,Oncology ,030220 oncology & carcinogenesis ,business ,Nasal cannula ,030215 immunology - Abstract
Patients with hematological malignancies are at risk for poor outcomes when diagnosed with Coronavirus Disease 2019 (COVID-19). It remains unclear whether cytopenias and specific leukemia subtypes play a role in the clinical course of COVID-19 infection. Here we report outcomes and their clinical/laboratory predictors for 65 patients with acute and chronic leukemias diagnosed with COVID-19 between March 8, 2020 and May 19, 2020 at Memorial Sloan Kettering Cancer Center in New York City. Most patients had CLL (48%) or AML (26%). A total of 14 (22%) patients required high flow nasal cannula or were intubated for mechanical ventilation and 11 patients (17%) died. A diagnosis of AML (OR 4.7, p=0.028), active treatment within the last 3 months (OR 5.22, p=0.047), neutropenia within seven days prior and up to 28 days after SARS-CoV-2 diagnosis (11.75, p=0.001) and ≥ 3 comorbidities (OR 6.55, p=0.019) were associated with increased odds of death.
- Published
- 2021
- Full Text
- View/download PDF
14. Interventions and outcomes of adult patients with B-ALL progressing after CD19 chimeric antigen receptor T-cell therapy
- Author
-
Brigitte Senechal, Mark B. Geyer, Marco L. Davila, Kevin J. Curran, Mithat Gonen, Mikhail Roshal, Isabelle Riviere, Michel Sadelain, Peter Maslak, Jessica Flynn, Kitsada Wudhikarn, Claudia Diamonte, Renier J. Brentjens, Jae H. Park, Xiuyan Wang, and Elizabeth Halton
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Immunology ,Psychological intervention ,Immunotherapy, Adoptive ,Biochemistry ,Disease-Free Survival ,CD19 ,Refractory ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Antibodies, Bispecific ,medicine ,Humans ,Inotuzumab Ozogamicin ,Aged ,Salvage Therapy ,Response rate (survey) ,biology ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Chimeric antigen receptor ,Survival Rate ,Natural history ,biology.protein ,Female ,Chimeric Antigen Receptor T-Cell Therapy ,Blinatumomab ,business ,human activities ,medicine.drug - Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has become a breakthrough treatment of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, despite the high initial response rate, the majority of adult patients with B-ALL progress after CD19 CAR T-cell therapy. Data on the natural history, management, and outcome of adult B-ALL progressing after CD19 CAR T cells have not been described in detail. Herein, we report comprehensive data of 38 adult patients with B-ALL who progressed after CD19 CAR T therapy at our institution. The median time to progression after CAR T-cell therapy was 5.5 months. Median survival after post–CAR T progression was 7.5 months. A high disease burden at the time of CAR T-cell infusion was significantly associated with risk of post–CAR T progression. Thirty patients (79%) received salvage treatment of post–CAR T disease progression, and 13 patients (43%) achieved complete remission (CR), but remission duration was short. Notably, 7 (58.3%) of 12 patients achieved CR after blinatumomab and/or inotuzumab administered following post–CAR T failure. Multivariate analysis revealed that a longer remission duration from CAR T cells was associated with superior survival after progression following CAR T-cell therapy. In summary, overall prognosis of adult B-ALL patients progressing after CD19 CAR T cells was poor, although a subset of patients achieved sustained remissions to salvage treatments, including blinatumomab, inotuzumab, and reinfusion of CAR T cells. Novel therapeutic strategies are needed to reduce risk of progression after CAR T-cell therapy and improve outcomes of these patients.
- Published
- 2021
- Full Text
- View/download PDF
15. Considerations for Use of Hematopoietic Growth Factors in Patients With Cancer Related to the COVID-19 Pandemic
- Author
-
Elizabeth A. Griffiths, Martha Wadleigh, Pamela S. Becker, Mark B. Geyer, Peter Westervelt, Avyakta Kallam, Sumithira Vasu, Rita Cool, Ivana Gojo, Rachel P. Rosovsky, Laura M Alwan, Anna Brown, Dwight D. Kloth, Lia Perez, Gary H. Lyman, Peter T. Curtin, Hope S. Rugo, Eric H. Kraut, Kimo Bachiashvili, Wajih Zaheer Kidwai, Sudipto Mukherjee, and Vivek Roy
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,Anemia ,medicine.medical_treatment ,Thrombopoietin mimetics ,Cancer ,Context (language use) ,Neutropenia ,medicine.disease ,Article ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Pandemic ,medicine ,030212 general & internal medicine ,business - Abstract
Hematopoietic growth factors, including erythrocyte stimulating agents (ESAs), granulocyte colony-stimulating factors, and thrombopoietin mimetics, can mitigate anemia, neutropenia, and thrombocytopenia resulting from chemotherapy for the treatment of cancer. In the context of pandemic SARS-CoV-2 infection, patients with cancer have been identified as a group at high risk of morbidity and mortality from this infection. Our subcommittee of the NCCN Hematopoietic Growth Factors Panel convened a voluntary group to review the potential value of expanded use of such growth factors in the current high-risk environment. Although recommendations are available on the NCCN website in the COVID-19 Resources Section (https://www.nccn.org/covid-19/), these suggestions are provided without substantial context or reference. Herein we review the rationale and data underlying the suggested alterations to the use of hematopoietic growth factors for patients with cancer in the COVID-19 era.
- Published
- 2021
- Full Text
- View/download PDF
16. An Ongoing Pilot Study of Targeted Radioimmunotherapy (131-I Apamistamab) Conditioning Prior to CD19-Targeted CAR T-Cell Therapy for Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia or Diffuse Large B-Cell Lymphoma
- Author
-
Mark B. Geyer, Briana Cadzin, Kinga Hosszu, Brigitte Senechal, Jennifer A. Spross, Alexis Mark, Avinash Desai, Elizabeth R. Cathcart, M. Lia Palomba, and Neeta Pandit-Taskar
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
17. Pediatric-inspired chemotherapy incorporating pegaspargase is safe and results in high rates of minimal residual disease negativity in adults up to age 60 with Philadelphia chromosome-negative acute lymphoblastic leukemia
- Author
-
Arati V. Rao, Martin S. Tallman, Jessica Flynn, Jose M. Salcedo, Peter Maslak, Qi Gao, Mikhail Roshal, Madhulika Shukla, Dan Douer, Sean M. Devlin, Mark B. Geyer, Meier Hsu, Jae H. Park, Ellen K. Ritchie, and Shreya Vemuri
- Subjects
Adult ,Asparaginase ,medicine.medical_specialty ,Neoplasm, Residual ,medicine.medical_treatment ,Context (language use) ,Gastroenterology ,Article ,Polyethylene Glycols ,Young Adult ,chemistry.chemical_compound ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Philadelphia Chromosome ,Child ,Pegaspargase ,Chemotherapy ,business.industry ,Lymphoblastic lymphoma ,Cancer ,Hematology ,Minimal Residual Disease Negativity ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Regimen ,chemistry ,business ,medicine.drug - Abstract
Administration of pediatric-inspired chemotherapy to adults up to age 60 with acute lymphoblastic leukemia (ALL) is challenging in part due to toxicities of asparaginase as well as myelosuppression. We conducted a multi-center phase II clinical trial (clinicaltrials gov. Identifier: NCT01920737) investigating a pediatric-inspired regimen, based on the augmented arm of the Children’s Cancer Group 1882 protocol, incorporating six doses of pegaspargase 2,000 IU/m2, rationally synchronized to avoid overlapping toxicity with other agents. We treated 39 adults aged 20-60 years (median age 38 years) with newly-diagnosed ALL (n=31) or lymphoblastic lymphoma (n=8). Grade 3-4 hyperbilirubinemia occurred frequently and at higher rates in patients aged 40-60 years (n=18) versus 18-39 years (n=21) (44% vs. 10%, P=0.025). However, eight of nine patients rechallenged with pegaspargase did not experience recurrent grade 3-4 hyperbilirubinemia. Grade 3-4 hypertriglyceridemia and hypofibrinogenemia were common (each 59%). Asparaginase activity at 7 days post-infusion reflected levels associated with adequate asparagine depletion, even among those with antibodies to pegaspargase. Complete response (CR)/CR with incomplete hematologic recovery was observed post-induction in 38 of 39 (97%) patients. Among patients with ALL, rates of minimal residual disease negativity by multi-parameter flow cytometry were 33% and 83% following induction phase I and phase II, respectively. Event-free and overall survival at 3 years (67.8% and 76.4%) compare favorably to outcomes observed in other series. These results demonstrate pegaspargase can be administered in the context of intensive multi-agent chemotherapy to adults aged ≤60 years with manageable toxicity. This regimen may serve as an effective backbone into which novel agents may be incorporated in future frontline studies. Trial registration: https://clinicaltrials. gov/ct2/show/NCT01920737
- Published
- 2020
- Full Text
- View/download PDF
18. Hypofibrinogenemia and disseminated intravascular coagulation rarely complicate treatment-naïve acute lymphoblastic leukemia
- Author
-
Simon Mantha, Martin S. Tallman, Jae H. Park, Charles Gaulin, Andriy Derkach, Angela Chan, and Mark B. Geyer
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lymphoblastic Leukemia ,Therapy naive ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Pathological ,Disseminated intravascular coagulation ,Acute leukemia ,business.industry ,Hematology ,Disseminated Intravascular Coagulation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Hypofibrinogenemia ,Afibrinogenemia ,medicine.disease ,Hyperfibrinolysis ,Oncology ,030220 oncology & carcinogenesis ,Hemostasis ,business ,circulatory and respiratory physiology ,030215 immunology - Abstract
Hypofibrinogenemia (HF) in acute leukemia has largely been attributed to hyperfibrinolysis and disseminated intravascular coagulation (DIC). Pathological alterations in hemostasis through various m...
- Published
- 2020
- Full Text
- View/download PDF
19. Dermatologic Adverse Events in Acute Lymphocytic Leukemia Patients Treated with Bispecific T-Cell Engager Blinatumomab
- Author
-
Rose Parisi, Emily A Cowen, Stephanie Gu, Melissa Pulitzer, Mark B. Geyer, Amber C. King, and Alina Markova
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
20. Outcomes of relapsed B-cell acute lymphoblastic leukemia after sequential treatment with blinatumomab and inotuzumab
- Author
-
Kitsada Wudhikarn, Amber C. King, Mark B. Geyer, Mikhail Roshal, Yvette Bernal, Boglarka Gyurkocza, Miguel-Angel Perales, and Jae H. Park
- Subjects
Adult ,Lymphoma, B-Cell ,Recurrence ,Antibodies, Bispecific ,Antigens, CD19 ,Humans ,Inotuzumab Ozogamicin ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Burkitt Lymphoma - Abstract
Novel monoclonal antibody (mAb)-based therapies targeting CD19 and CD22 (blinatumomab and inotuzumab) have shown high rates of complete remission (CR) and been used as a bridging treatment to potentially curative allogeneic hematopoietic stem cell transplantation (alloHSCT) in adults with relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). However, limited data exist on the outcome of patients resistant to both mAbs as well as responses to each agent when progressed after the alternate antigen-targeted mAb. Herein, we report outcomes of 29 patients with R/R B-ALL previously treated with both blinatumomab and inotuzumab. Twenty-five patients (86.2%) received blinatumomab as first mAb (mAb1), and CD19-negative/dim relapses were observed in 44% of the patients. Inotuzumab induced CR in 68% of the patients for post-blinatumomab relapse regardless of CD19 expression status. The median time between mAb1 and mAb2 was 99 days. Twelve (63.2%) of 19 patients who achieved remission after mAb2 underwent alloHSCT. The median time from mAb2 to alloHSCT was 37.5 days. Acute graft-versus-host disease and nonrelapse mortality were observed in 58.3% (grade 3 or higher, 25%) and 41.7%, respectively. With a median follow-up of 16.8 months after mAb2, 19 patients (65.5%) relapsed, and 21 patients (72.4%) have died. Overall survival was not different between alloHSCT and non-alloHSCT patients. In conclusion, patients with B-ALL who relapsed after blinatumomab could be successfully rescued by inotuzumab as a bridge to alloHSCT but represent an ultra-high-risk group with poor overall survival. Further studies, including novel consolidation and treatment sequence, may improve outcomes of these patients.
- Published
- 2021
21. A phase 2 biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas
- Author
-
Brielle Liotta, Natasha Galasso, Lauren Pomerantz, Jürgen Rademaker, Esther Drill, Ahmet Dogan, Jeeyeon Baik, Heiko Schöder, Steven M. Horwitz, Leslie Perez, William Blouin, Theresa Davey, Obadi Obadi, Giorgio Inghirami, Eric D. Jacobsen, Jonathan H. Schatz, Mark B. Geyer, Ariela Noy, Jack Dowd, Santosha A. Vardhana, David J. Straus, Nancy Yi, Sarah J. Noor, David M. Weinstock, Travis J. Hollmann, Helen Hancock, Priyadarshini Kumar, Nivetha Ganesan, Paola Ghione, Jia Ruan, Alison J. Moskowitz, Rachel Neuman, Alayna Santarosa, Sunyoung Ryu, Patricia L. Myskowski, and Samia Sohail
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Ruxolitinib ,Large granular lymphocytic leukemia ,Immunology ,Phases of clinical research ,Biochemistry ,Young Adult ,Internal medicine ,Nitriles ,medicine ,Clinical endpoint ,Humans ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Aged ,Janus Kinases ,Aged, 80 and over ,Mycosis fungoides ,Lymphoid Neoplasia ,Hematology ,business.industry ,Lymphoma, T-Cell, Peripheral ,Cell Biology ,Middle Aged ,medicine.disease ,Lymphoma ,STAT Transcription Factors ,Pyrimidines ,Treatment Outcome ,Biomarker (medicine) ,Pyrazoles ,Female ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Signaling through JAK1 and/or JAK2 is common among tumor and nontumor cells within peripheral T-cell lymphoma (PTCL). No oral therapies are approved for PTCL, and better treatments for relapsed/refractory disease are urgently needed. We conducted a phase 2 study of the JAK1/2 inhibitor ruxolitinib for patients with relapsed/refractory PTCL (n = 45) or mycosis fungoides (MF) (n = 7). Patients enrolled onto 1 of 3 biomarker-defined cohorts: (1) activating JAK and/or STAT mutations, (2) ≥30% pSTAT3 expression among tumor cells by immunohistochemistry, or (3) neither or insufficient tissue to assess. Patients received ruxolitinib 20 mg PO twice daily until progression and were assessed for response after cycles 2 and 5 and every 3 cycles thereafter. The primary endpoint was clinical benefit rate (CBR), defined as the combination of complete response, partial response (PR), and stable disease lasting at least 6 months. Only 1 of 7 patients with MF had CBR (ongoing PR > 18 months). CBR among the PTCL cases (n = 45) in cohorts 1, 2, and 3 were 53%, 45%, and 13% (cohorts 1 & 2 vs 3, P = .02), respectively. Eight patients had CBR > 12 months (5 ongoing), including 4 of 5 patients with T-cell large granular lymphocytic leukemia. In an exploratory analysis using multiplex immunofluorescence, expression of phosphorylated S6, a marker of PI3 kinase or mitogen-activated protein kinase activation, in
- Published
- 2021
22. Molecular Predictors and Effectiveness of Measurable Residual Disease (MRD) Eradication with Chemotherapy and Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia
- Author
-
Maria E. Arcila, Omar Abdel-Wahab, Miguel-Angel Perales, Kamal Menghrajani, Maximilian Stahl, Mikhail Roshal, Christina Cho, Elli Papaemmanuil, Juliet N. Barker, Jacob L. Glass, Sergio Giralt, Sean M. Devlin, Justin Taylor, Noushin Farnoud, Andrew Dunbar, Martin S. Tallman, Brian C. Shaffer, Andriy Derkach, Bartlomiej Getta, Esperanza B. Papadopoulos, Christopher Famulare, Eytan M. Stein, Boglarka Gyurkocza, Ross L. Levine, Aaron D. Viny, Sheng F. Cai, Jessica Schulman, Erin McGovern, Yanming Zhang, Minal Patel, Zachary D. Epstein-Peterson, Mark B. Geyer, and Aaron D Goldberg
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Disease ,Biochemistry ,Transplantation ,Internal medicine ,Medicine ,Stem cell ,business - Abstract
Background: Measurable residual disease (MRD) is a powerful prognostic factor in AML, including in prediction of outcomes post allogeneic stem cell transplant (alloSCT). However, genomic predictors of successful MRD eradication with chemotherapy prior to alloSCT are unclear. Objectives: Here we provide an integrated analysis of 233 patients (pts) who underwent induction chemotherapy with baseline next-generation sequencing (NGS) followed by serial immunophenotypic monitoring for MRD while patients received additional therapy and alloSCT. Methods: All pts who received anthracycline + cytarabine, +/- investigational agents at Memorial Sloan Kettering Cancer Center starting in April 2014 were retrospectively studied (A). 142 out of 233 pts subsequently underwent alloSCT after induction or additional therapy (A). Immunophenotypic MRD was identified in bone marrow aspirates (BMA) by multiparameter flow cytometry. Any level of residual disease was considered MRD+. Molecular analysis was obtained from pre-induction BMA by NGS using 28 or 49 or 400 gene panels. Results: Patient and treatment characteristics for all pts are detailed in panel (B). Induction chemotherapy resulted in an MRD-CR/CRi and MRD+CR/CRi in 29% and 23% of all pts, respectively (C). Additional therapy included consolidation (n=51), intensive re-induction/salvage (n=47) and non-intensive therapy (n=9). Of 83 AML pts with persistent AML and 58 pts with MRD+CR/CRi after induction (R1), 38/141 (27%) were able to be converted to MRD-CR/CRi. While 33/38 of pts went on to alloSCT after conversion to MRD-CR/CRi, 22 and 36 pts went to alloSCT with persistent AML and MRD+CR/CRi AML, respectively. We focused on pre-induction molecular predictors for achieving an MRD-CR/CRi response prior to transplant for the 142 pts who underwent alloSCT (D). Pts with a NPM1 (79%, Odds ratio [OR] 3.7, p=0.01), IDH1 (92%, OR 3.9, p=0.01) and KRAS (100%, OR 5.0, p=0.03) mutations achieved high rates of MRD-CR/CRi prior to alloSCT. In contrast, RUNX1 (28%, OR 0.2, p=0.01), TP53 (12%, OR 0.1, p=0.02) and SF3B1 (14%, OR 0.1, p=0.04) mutations predicted decreased odds of achieving MRD-CR/CRi prior to alloSCT despite induction and post-induction therapy. AlloSCT resulted in high rates of conversion from MRD+ and persistent disease to MRD negativity. Most pts who entered transplant with CR/CRi MRD+ (28/36, 76%) or persistent AML (14/22, 64%) cleared MRD by the first post-transplant BMA at a median of 32 days (E). Post-alloSCT follow-up indicated value in converting MRD+ to MRD- prior to alloSCT. There was no significant difference in post-transplant cumulative incidence of relapse (F) and OS (G) between early MRD-CR/CRi immediately following induction versus later conversion to MRD-CR/CRi with additional therapy prior to alloSCT. Despite initial post-transplant MRD clearance, pts who entered alloSCT with persistent AML or MRD+ had higher incidence of relapse (p=0.00037, F) and poorer post-transplant OS (p=0.013, G) compared to pts who entered alloSCT with MRD-. Pts with persistent disease prior to alloSCT had shorter duration of MRD- induced by alloSCT compared to pts with MRD-CR/CRi after induction or converted MRD-CR/CRi prior to alloSCT (p=0.0042, H). Importantly, duration of MRD negativity after alloSCT for patients who achieved MRD- prior to alloSCT was not affected by whether patients received induction +/- consolidation (I: treatment type 1-3 from B) vs. induction and salvage treatment for refractory AML (I: treatment type 4-6 from B). Conclusion: We show that transplanted AML pts with specific molecular mutations (RUNX1, SF3B1, and TP53) are unlikely to achieve MRD-CR/CRi after induction, consolidation or salvage therapy, while other mutations (NPM1, IDH1, KRAS) predict high rates of MRD- prior to alloSCT. Additional post-induction therapy may be advantageous for some MRD+ pts to achieve MRD- prior to alloSCT. Post-transplant OS is improved in pts who are MRD- at time of transplant, regardless of whether they required additional therapy beyond induction to achieve this state. AlloSCT is highly effective at eradicating MRD, but post-transplant MRD- is more durable in pts who are MRD- pre-alloSCT. Our results suggest that development of MRD-eradicating therapies has the potential to improve post-transplant outcomes and argues for innovative trials for pts with adverse molecular features currently unlikely to achieve MRD- pre alloSCT. Figure Disclosures Cai: Imago Biosciences, Inc.: Consultancy, Current equity holder in private company; DAVA Oncology: Honoraria. Geyer:Amgen: Research Funding. Glass:Gerson Lehman Group: Consultancy. Stein:Syros: Membership on an entity's Board of Directors or advisory committees; PTC Therapeutics: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Biotheryx: Consultancy; Bayer: Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Syndax: Consultancy, Research Funding; Seattle Genetics: Consultancy; Abbvie: Consultancy; Amgen: Consultancy; Celgene Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Agios Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Levine:Gilead: Honoraria; Isoplexis: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Qiagen: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Research Funding; Janssen: Consultancy; Lilly: Consultancy, Honoraria; Imago: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; C4 Therapeutics: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy; Novartis: Consultancy; Prelude Therapeutics: Research Funding; Loxo: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria; Morphosys: Consultancy; Roche: Consultancy, Honoraria, Research Funding. Gyurkocza:Actinium: Research Funding. Perales:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Nektar Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; MolMed: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; Medigene: Membership on an entity's Board of Directors or advisory committees, Other; Servier: Membership on an entity's Board of Directors or advisory committees, Other; Omeros: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Consultancy, Honoraria; NexImmune: Membership on an entity's Board of Directors or advisory committees; Cidara Therapeutics: Other; Miltenyi Biotec: Research Funding; Kite/Gilead: Honoraria, Research Funding; Incyte Corporation: Honoraria, Research Funding; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria; Bellicum: Honoraria, Membership on an entity's Board of Directors or advisory committees. Abdel-Wahab:H3 Biomedicine Inc.: Consultancy, Research Funding; Janssen: Consultancy; Envisagenics Inc.: Current equity holder in private company; Merck: Consultancy. Papaemmanuil:Kyowa Hakko Kirin: Consultancy, Honoraria; Isabl: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; MSKCC: Patents & Royalties; Novartis: Consultancy, Honoraria; Illumina: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Prime Oncology: Consultancy, Honoraria. Giralt:KITE: Consultancy; NOVARTIS: Consultancy, Honoraria, Research Funding; OMEROS: Consultancy, Honoraria; AMGEN: Consultancy, Research Funding; TAKEDA: Research Funding; ACTINUUM: Consultancy, Research Funding; MILTENYI: Consultancy, Research Funding; CELGENE: Consultancy, Honoraria, Research Funding; JAZZ: Consultancy, Honoraria. Tallman:Glycomimetics: Research Funding; Rafael: Research Funding; Amgen: Research Funding; Bioline rx: Membership on an entity's Board of Directors or advisory committees; Daiichi-Sankyo: Membership on an entity's Board of Directors or advisory committees; KAHR: Membership on an entity's Board of Directors or advisory committees; UpToDate: Patents & Royalties; Rigel: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees; Jazz Pharma: Membership on an entity's Board of Directors or advisory committees; Oncolyze: Membership on an entity's Board of Directors or advisory committees; Delta Fly Pharma: Membership on an entity's Board of Directors or advisory committees; BioSight: Membership on an entity's Board of Directors or advisory committees, Research Funding; ADC Therapeutics: Research Funding; Orsenix: Research Funding; Cellerant: Research Funding; Abbvie: Research Funding. Goldberg:AROG: Research Funding; Aprea: Research Funding; ADC Therapeutics: Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy; Aptose: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Dava Oncology: Honoraria; Pfizer: Research Funding; Celularity: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
23. BRAF in the cross-hairs
- Author
-
Mark B. Geyer, Martin S. Tallman, and Omar Abdel-Wahab
- Subjects
Proto-Oncogene Proteins B-raf ,Oncology ,medicine.medical_specialty ,endocrine system diseases ,MAP Kinase Signaling System ,medicine.medical_treatment ,Lymphoproliferative disorders ,Antineoplastic Agents ,Article ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Internal medicine ,medicine ,Animals ,Humans ,Point Mutation ,Hairy cell leukemia ,Vemurafenib ,Adverse effect ,Protein Kinase Inhibitors ,neoplasms ,Leukemia, Hairy Cell ,business.industry ,Clinical course ,Hematology ,medicine.disease ,Activating mutation ,digestive system diseases ,030220 oncology & carcinogenesis ,business ,Signal Transduction ,030215 immunology ,medicine.drug - Abstract
INTRODUCTION Hairy cell leukemia (HCL) is a rare, chronic B-cell lymphoproliferative disorder characterized by distinctive morphologic features and an indolent clinical course. The discovery of a recurrent activating mutation in BRAF (BRAF V600E) as a disease-defining genetic event in HCL has substantial diagnostic and therapeutic implications. Areas covered: Herein the authors review the role of BRAF V600E and RAF-MEK-ERK signaling in the pathogenesis of HCL, anecdotal clinical reports of BRAF inhibitor monotherapy in management of relapsed or refractory HCL, larger phase 2 trials investigating efficacy of BRAF inhibitor therapy for HCL, adverse effects commonly associated with BRAF inhibitor therapy, including cutaneous toxicity, and mechanisms of therapeutic resistance. Expert opinion: Ongoing and planned studies will help to optimize the use of BRAF inhibitor therapy for HCL by determining the efficacy of BRAF inhibition in combination with other antigen targeted or molecularly targeted therapies, and more broadly, to determine how hematologists can best utilize and sequence emerging diagnostic and therapeutic modalities in the care of patients with newly diagnosed and relapsed or refractory HCL.
- Published
- 2019
- Full Text
- View/download PDF
24. Tolerability and toxicity of pegaspargase in adults 40 years and older with acute lymphoblastic leukemia
- Author
-
Charlene C. Kabel, Mark B. Geyer, Sridevi Rajeeve, Jeremy J. Pappacena, Sarah E. Stump, Martin S. Tallman, Jessica A. Lavery, Ryan J. Daley, and Jae H. Park
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Asparaginase ,Lymphoblastic Leukemia ,Antineoplastic Agents ,Polyethylene Glycols ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Asparagine ,Pegaspargase ,business.industry ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Leukemia ,chemistry ,Tolerability ,030220 oncology & carcinogenesis ,Toxicity ,business ,030215 immunology ,medicine.drug - Abstract
Pegaspargase is a modified version of asparaginase with prolonged asparagine depletion. It appears to be safe in adults40 years old, but has a unique spectrum of toxicities, the risks of which appear to increase with age. The primary objective of this study was to evaluate pegaspargase tolerability and toxicity as assessed by evaluation of incidence and severity of adverse events. Secondary objectives included characterization of the reasons underlying pegaspargase discontinuation, when applicable. Grade 3/4 asparaginase-related toxicities with ≥10% incidence included: hyperbilirubinemia, hyperglycemia, hypertriglyceridemia, hypoalbuminemia, hypofibrinogenemia, and transaminitis. 63% of patients (38 of 60) received all intended doses of pegaspargase, with the most common reasons for discontinuation noted as hypersensitivity (12%), hyperbilirubinemia/transaminitis (8%), and hematopoietic transplantation in complete remission (10%). This study suggests that while hepatotoxicity and other known adverse effects are common, with careful monitoring, pegaspargase can safely be administered to adults with ALL age ≥40 years old.
- Published
- 2020
25. FGFR1 Rearrangement Guides Diagnosis and Treatment of a Trilineage B, T, and Myeloid Mixed Phenotype Acute Leukemia
- Author
-
Rayma Benayed, Qi Gao, Ahmet Dogan, Mark B. Geyer, Wenbin Xiao, Ramya Gadde, Xiaoli Mi, Yanming Zhang, Ying Liu, Maria E. Arcila, and Mikhail Roshal
- Subjects
Cancer Research ,Myeloid ,medicine.anatomical_structure ,Mixed phenotype acute leukemia ,Text mining ,Oncology ,business.industry ,Fibroblast growth factor receptor 1 ,medicine ,Cancer research ,Case Reports ,business - Published
- 2020
26. Outcomes of COVID-19 in patients with CLL: a multicenter international experience
- Author
-
Neil Bailey, Fatima Miras, Jose Angel Hernandez-Rivas, Chadi Nabhan, John M. Pagel, Elise A. Chong, Manali Kamdar, Sigrid S. Skånland, Raul Cordoba, Matthew S. Davids, Mazyar Shadman, Angus Broom, Ellin Berman, Shuo Ma, Anthony R. Mato, Paul M. Barr, Meera Patel, Lindsey E. Roeker, Erlene K. Seymour, José A. García-Marco, Andrew D. Zelenetz, Anders Österborg, Matthew R. Wilson, Toby A. Eyre, Danielle M. Brander, Krista Isaac, Jeffrey Pu, Mark B. Geyer, Richard R. Furman, Sonia Lebowitz, Renata Walewska, Talha Munir, Nikita Malakhov, John N. Allan, Scott F. Huntington, Inhye E. Ahn, Darko Antic, Lotta Hanson, Adrian Wiestner, Ryan Jacobs, Paola Ghione, Nicolas Martinez-Calle, Lori A. Leslie, Erica Bhavsar, Suchitra Sundaram, Daniel Wojenski, Jennifer R. Brown, Chaitra S. Ujjani, Amanda N. Seddon, Daniel Naya, Javier López-Jiménez, Harriet S. Walter, Christine E. Ryan, Craig A. Portell, Krish Patel, Dima El-Sharkawi, Michael Koropsak, Guilherme Fleury Perini, Noemi Fernandez Escalada, Helen Parry, Nicole Lamanna, and Piers E.M. Patten
- Subjects
0301 basic medicine ,Male ,Clinical Trials and Observations ,Chronic lymphocytic leukemia ,Anti-Inflammatory Agents ,Disease ,Biochemistry ,law.invention ,0302 clinical medicine ,law ,Case fatality rate ,Agammaglobulinaemia Tyrosine Kinase ,Aged, 80 and over ,Risk of infection ,Hematology ,Middle Aged ,Intensive care unit ,3. Good health ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,Coronavirus Infections ,BLOOD Commentary ,Adult ,medicine.medical_specialty ,Immunology ,Pneumonia, Viral ,Antiviral Agents ,03 medical and health sciences ,Betacoronavirus ,Internal medicine ,medicine ,Humans ,Pandemics ,Protein Kinase Inhibitors ,Survival analysis ,COVID-19 Serotherapy ,Aged ,business.industry ,SARS-CoV-2 ,Immunization, Passive ,COVID-19 ,Cell Biology ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Analysis ,Clinical trial ,Pneumonia ,030104 developmental biology ,business - Abstract
There is a Blood Commentary on this article in this issue., Key Points Both watch-and-wait and treated CLL patients have high mortality rates when admitted for COVID-19. Receiving a BTKi for CLL at COVID-19 diagnosis severe enough to require hospitalization did not influence case fatality rate in this study., Given advanced age, comorbidities, and immune dysfunction, chronic lymphocytic leukemia (CLL) patients may be at particularly high risk of infection and poor outcomes related to coronavirus disease 2019 (COVID-19). Robust analysis of outcomes for CLL patients, particularly examining effects of baseline characteristics and CLL-directed therapy, is critical to optimally manage CLL patients through this evolving pandemic. CLL patients diagnosed with symptomatic COVID-19 across 43 international centers (n = 198) were included. Hospital admission occurred in 90%. Median age at COVID-19 diagnosis was 70.5 years. Median Cumulative Illness Rating Scale score was 8 (range, 4-32). Thirty-nine percent were treatment naive (“watch and wait”), while 61% had received ≥1 CLL-directed therapy (median, 2; range, 1-8). Ninety patients (45%) were receiving active CLL therapy at COVID-19 diagnosis, most commonly Bruton tyrosine kinase inhibitors (BTKi’s; n = 68/90 [76%]). At a median follow-up of 16 days, the overall case fatality rate was 33%, though 25% remain admitted. Watch-and-wait and treated cohorts had similar rates of admission (89% vs 90%), intensive care unit admission (35% vs 36%), intubation (33% vs 25%), and mortality (37% vs 32%). CLL-directed treatment with BTKi’s at COVID-19 diagnosis did not impact survival (case fatality rate, 34% vs 35%), though the BTKi was held during the COVID-19 course for most patients. These data suggest that the subgroup of CLL patients admitted with COVID-19, regardless of disease phase or treatment status, are at high risk of death. Future epidemiologic studies are needed to assess severe acute respiratory syndrome coronavirus 2 infection risk, these data should be validated independently, and randomized studies of BTKi’s in COVID-19 are needed to provide definitive evidence of benefit., Visual Abstract
- Published
- 2020
27. Allogeneic stem cell transplantation for chronic lymphocytic leukemia in the era of novel agents
- Author
-
Matthew S. Davids, Timothy J Voorhees, Anthony R. Mato, Toby A. Eyre, Haesook T. Kim, Craig S. Sauter, Gemlyn George, Sergio Giralt, Miguel-Angel Perales, Catherine C. Coombs, Harriet S. Walter, Jae H. Park, Lindsey E. Roeker, Andrew D. Zelenetz, Oscar B Lahoud, Steven T. Manchini, Danielle M. Brander, Alan P Skarbnik, Mark B. Geyer, Andrea Sitlinger, Peter Dreger, Kim Orchard, Nirav N. Shah, Jennifer R. Brown, and Arvind Arumainathan
- Subjects
Oncology ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,chemistry.chemical_compound ,Chemoimmunotherapy ,Internal medicine ,medicine ,Humans ,Cumulative incidence ,Lymphoma, Follicular ,Retrospective Studies ,Lymphoid Neoplasia ,Venetoclax ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Transplantation ,Graft-versus-host disease ,chemistry ,Ibrutinib ,business - Abstract
Although novel agents (NAs) have improved outcomes for patients with chronic lymphocytic leukemia (CLL), a subset will progress through all available NAs. Understanding outcomes for potentially curative modalities including allogeneic hematopoietic stem cell transplantation (alloHCT) following NA therapy is critical while devising treatment sequences aimed at long-term disease control. In this multicenter, retrospective cohort study, we examined 65 patients with CLL who underwent alloHCT following exposure to ≥1 NA, including baseline disease and transplant characteristics, treatment preceding alloHCT, transplant outcomes, treatment following alloHCT, and survival outcomes. Univariable and multivariable analyses evaluated associations between pre-alloHCT factors and progression-free survival (PFS). Twenty-four-month PFS, overall survival (OS), nonrelapse mortality, and relapse incidence were 63%, 81%, 13%, and 27% among patients transplanted for CLL. Day +100 cumulative incidence of grade III-IV acute graft-vs-host disease (GVHD) was 24%; moderate-severe GVHD developed in 27%. Poor-risk disease characteristics, prior NA exposure, complete vs partial remission, and transplant characteristics were not independently associated with PFS. Hematopoietic cell transplantation–specific comorbidity index independently predicts PFS. PFS and OS were not impacted by having received NAs vs both NAs and chemoimmunotherapy, 1 vs ≥2 NAs, or ibrutinib vs venetoclax as the line of therapy immediately pre-alloHCT. AlloHCT remains a viable long-term disease control strategy that overcomes adverse CLL characteristics. Prior NAs do not appear to impact the safety of alloHCT, and survival outcomes are similar regardless of number of NAs received, prior chemoimmunotherapy exposure, or NA immediately preceding alloHCT. Decisions about proceeding to alloHCT should consider comorbidities and anticipated response to remaining therapeutic options.
- Published
- 2020
28. Contributors
- Author
-
James L. Abbruzzese, Omar Abdel-Wahab, Ghassan K. Abou-Alfa, Janet L. Abrahm, Jeffrey S. Abrams, Jeremy S. Abramson, Dara L. Aisner, Michelle Alonso-Basanta, Jesus Anampa, Megan E. Anderson, Emmanuel S. Antonarakis, Richard Aplenc, Frederick R. Appelbaum, Luiz H. Araujo, Ammar Asban, Edward Ashwood, Farrukh T. Awan, Juliet L. Aylward, Arjun V. Balar, Courtney J. Balentine, Stefan K. Barta, Nancy Bartlett, Karen Basen-Engquist, Lynda Kwon Beaupin, Ross S. Berkowitz, Donald A. Berry, Therese Bevers, John F. Boggess, Julie R. Brahmer, Janet Brown, Karen Brown, Powel Brown, Ilene Browner, Paul A. Bunn, William R. Burns, John C. Byrd, Karen Cadoo, David P. Carbone, H. Ballentine Carter, Jorge J. Castillo, Alfred E. Chang, Eric Chang, Stephen J. Chanock, Claudia I. Chapuy, Vikash P. Chauhan, Herbert Chen, Ronald C. Chen, Nai-Kong V. Cheung, Jennifer H. Choe, Michaele C. Christian, Paul M. Cinciripini, Michael F. Clarke, Robert E. Coleman, Robert L. Coleman, Adriana M. Coletta, Jerry M. Collins, Jean M. Connors, Michael Cools, Kevin R. Coombes, Jorge Cortes, Mauro W. Costa, Anne Covey, Kenneth H. Cowan, Christopher H. Crane, Jeffrey Crawford, Kristy Crooks, Daniel J. Culkin, Brian G. Czito, Piero Dalerba, Josep Dalmau, Mai Dang, Michael D'Angelica, Kurtis D. Davies, Myrtle Davis, Nicolas Dea, Ana De Jesus-Acosta, Angelo M. DeMarzo, Theodore L. DeWeese, Maximilian Diehn, Subba R. Digumarthy, Angela Dispenzieri, Khanh T. Do, Konstantin Dobrenkov, Jeffrey S. Dome, James H. Doroshow, Jay F. Dorsey, Marianne Dubard-Gault, Steven G. DuBois, Dan G. Duda, Malcolm Dunlop, Linda R. Duska, Madeleine Duvic, Imane El Dika, Hashem El-Serag, Jeffrey M. Engelmann, David S. Ettinger, Lola A. Fashoyin-Aje, Eric R. Fearon, James M. Ford, Wilbur A. Franklin, Phoebe E. Freer, Boris Freidlin, Alison G. Freifeld, Terence W. Friedlander, Debra L. Friedman, Arian F. Fuller, Lorenzo Galluzzi, Mark C. Gebhardt, Daniel J. George, Mark B. Geyer, Amato J. Giaccia, Mark R. Gilbert, Whitney Goldner, Donald P. Goldstein, Annekathryn Goodman, Karyn A. Goodman, Kathleen Gordon, Laura Graeff-Armas, Alexander J. Greenstein, Stuart A. Grossman, Stephan Grupp, Arjun Gupta, Irfanullah Haider, Missak Haigentz, John D. Hainsworth, Benjamin E. Haithcock, Christopher L. Hallemeier, Samir Hanash, Aphrothiti J. Hanrahan, James Harding, Michael R. Harrison, Muneer G. Hasham, Ernest Hawk, Jonathan Hayman, Jonathan E. Heinlen, N. Lynn Henry, Joseph Herman, Brian P. Hobbs, Ingunn Holen, Leora Horn, Neil S. Horowitz, Steven M. Horwitz, Odette Houghton, Scott C. Howard, Clifford A. Hudis, Stephen P. Hunger, Arti Hurria, David H. Ilson, Annie Im, Gopa Iyer, Elizabeth M. Jaffee, Reshma Jagsi, Rakesh K. Jain, William Jarnagin, Aminah Jatoi, Anuja Jhingran, David H. Johnson, Brian Johnston, Patrick G. Johnston, Kevin D. Judy, Lisa A. Kachnic, Orit Kaidar-Person, Sanjeeva Kalva, Deborah Y. Kamin, Hagop Kantarjian, Giorgos Karakousis, Maher Karam-Hage, Nadine M. Kaskas, Michael B. Kastan, Nora Katabi, Daniel R. Kaul, Scott R. Kelley, Nancy Kemeny, Erin E. Kent, Oliver Kepp, Simon Khagi, Joshua E. Kilgore, D. Nathan Kim, Bette K. Kleinschmidt-DeMasters, Edward L. Korn, Guido Kroemer, Geoffrey Y. Ku, Shivaani Kummar, Bonnie Ky, Daniel A. Laheru, Paul F. Lambert, Mark Lawler, Jennifer G. Le-Rademacher, John Y.K. Lee, Nancy Y. Lee, Susanna L. Lee, Jonathan E. Leeman, Andreas Linkermann, Jinsong Liu, Simon Lo, Jason W. Locasale, Charles L. Loprinzi, Maeve Lowery, Emmy Ludwig, Matthew A. Lunning, Robert A. Lustig, Mitchell Machtay, Crystal Mackall, David A. Mahvi, David M. Mahvi, Amit Maity, Neil Majithia, Marcos Malumbres, Karen Colbert Maresso, John D. Martin, Koji Matsuo, Natalie H. Matthews, Lauren Mauro, R. Samuel Mayer, Worta McCaskill-Stevens, Megan A. McNamara, Neha Mehta-Shah, Robert E. Merritt, Matthew I. Milowsky, Lori M. Minasian, Tara C. Mitchell, Demytra Mitsis, Michelle Mollica, Margaret Mooney, Farah Moustafa, Lida Nabati, Jarushka Naidoo, Amol Narang, Heidi Nelson, William G. Nelson, Suzanne Nesbit, Mark Niglas, Tracey O'Connor, Kenneth Offit, Mihaela Onciu, Eileen M. O’Reilly, Elaine A. Ostrander, Lisa Pappas-Taffer, Drew Pardoll, Jae H. Park, Anery Patel, Anish J. Patel, Steven R. Patierno, Steven Z. Pavletic, Peter C. Phillips, Miriam D. Post, Amy A. Pruitt, Christiane Querfeld, Vance A. Rabius, S. Vincent Rajkumar, Mohammad O. Ramadan, Erinn B. Rankin, Sushanth Reddy, Michael A. Reid, Scott Reznik, Tina Rizack, Jason D. Robinson, Leslie Robinson-Bostom, Carlos Rodriguez-Galindo, Paul B. Romesser, Steven T. Rosen, Myrna R. Rosenfeld, Nadia Rosenthal, Meredith Ross, Julia H. Rowland, Anthony H. Russell, Michael S. Sabel, Arjun Sahgal, Ryan D. Salinas, Erin E. Salo-Mullen, Manuel Salto-Tellez, Sydney M. Sanderson, John T. Sandlund, Victor M. Santana, Michelle Savage, Eric C. Schreiber, Lynn Schuchter, Liora Schultz, Michael V. Seiden, Morgan M. Sellers, Payal D. Shah, Jinru Shia, Konstantin Shilo, Eric Small, Angela B. Smith, Stephen N. Snow, David B. Solit, Anil K. Sood, Enrique Soto-Perez-de-Celis, Joseph A. Sparano, Vladimir S. Spiegelman, Sheri L. Spunt, Zsofia K. Stadler, David P. Steensma, Richard M. Stone, Steven Kent Stranne, Kelly Stratton, Bill Sugden, Andrew M. Swanson, Martin S. Tallman, James E. Talmadge, David T. Teachey, Catalina V. Teba, Ayalew Tefferi, Bin Tean Teh, Joyce M.C. Teng, Joel E. Tepper, Premal H. Thaker, Aaron P. Thrift, Arthur-Quan Tran, Grace Triska, Donald Trump, Kenneth Tsai, Chia-Lin Tseng, Diane Tseng, Sandra Van Schaeybroeck, Brian A. Van Tine, Erin R. Vanness, Gauri Varadhachary, Marileila Varella-Garcia, Richard L. Wahl, Michael F. Walsh, Thomas Wang, Jared Weiss, Irving L. Weissman, Shannon N. Westin, Jeffrey D. White, Richard Wilson, Richard J. Wong, Gary S. Wood, Yaohui G. Xu, Meng Xu-Welliver, Shlomit Yust-Katz, Timothy Zagar, Elaine M. Zeman, Tian Zhang, and James A. Zwiebel
- Published
- 2020
- Full Text
- View/download PDF
29. Hairy Cell Leukemia
- Author
-
Mark B. Geyer, Omar Abdel-Wahab, Martin S. Tallman, and Jae H. Park
- Published
- 2020
- Full Text
- View/download PDF
30. Iomab with Adoptive Cellular Therapy (Iomab-ACT): A Pilot Study of 131-I Apamistamab Followed By CD19-Targeted CAR T-Cell Therapy for Patients with Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia or Diffuse Large B-Cell Lymphoma
- Author
-
Mark B. Geyer, Eileen M. Geoghegan, Briana R. Cadzin, Dale L. Ludwig, Neeta Pandit-Taskar, Peter Kane, Brigitte Senechal, Mark S. Berger, Vijay Reddy, Craig S. Sauter, and Elizabeth Halton
- Subjects
biology ,business.industry ,Immunology ,Cell Biology ,Hematology ,B-cell acute lymphoblastic leukemia ,medicine.disease ,Biochemistry ,CD19 ,Refractory ,Cancer research ,biology.protein ,Medicine ,Adoptive cellular therapy ,CAR T-cell therapy ,business ,Diffuse large B-cell lymphoma - Abstract
Background: Autologous CD19-targeted chimeric antigen receptor-modified (CAR) T-cell therapy leads to complete responses (CR) in patients (pts) with (w/) relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL, >80% CR rate) and diffuse large B-cell lymphoma (DLBCL, ~40-55% CR rate). However, following fludarabine/cyclophosphamide (Flu/Cy) conditioning and CAR T-cell therapy w/ a CD28 costimulatory domain (e.g. 19-28z CAR T-cells), rates of grade ≥3 ICANS and grade ≥3 cytokine release syndrome (CRS) in pts w/ R/R DLBCL and morphologic R/R B-ALL exceed 30%. CRS and ICANS are associated w/ considerable morbidity, including increased length of hospitalization, and may be fatal. Host monocytes appear to be the major reservoir of cytokines driving CRS and ICANS post-CAR T-cell therapy (Giavradis et al. and Norelli et al., Nature Medicine, 2018). Circulating monocytic myeloid-derived suppressor cells (MDSCs) may also blunt efficacy of 19-28z CAR T-cells in R/R DLBCL (Jain et al., Blood, 2021). The CD45-targeted antibody radioconjugate (ARC) 131-I apamistamab is being investigated at myeloablative doses as conditioning prior to hematopoietic cell transplantation in pts w/ R/R acute myeloid leukemia. However, even at low doses (4-20 mCi), transient lymphocyte and blast reduction are observed. Preclinical studies in C57BL/6 mice demonstrate low-dose anti CD45 radioimmunotherapy (100 microCi) transiently depletes >90% lymphocytes, including CD4/CD8 T-cells, B-cells, NK cells, and T-regs, as well as splenocytes and MDSCs, w/ negligible effect on bone marrow (BM) hematopoietic stem cells (Dawicki et al., Oncotarget, 2020). We hypothesized a higher, yet nonmyeloablative dose of 131-I apamistamab may achieve more sustained, but reversible depletion of lymphocytes and other CD45 + immune cells, including monocytes thought to drive CRS/ICANS. We additionally hypothesized this approach (vs Flu/Cy) prior to CAR T-cell therapy would promote CAR T-cell expansion while reducing CSF levels of monocyte-derived cytokines (e.g. IL-1, IL-6, and IL-10), thus lowering the risk of severe ICANS (Fig 1A). Study design and methods: We are conducting a single-institution pilot study of 131-I apamistamab in lieu of Flu/Cy prior to 19-28z CAR T-cells in adults w/ R/R BALL or DLBCL (NCT04512716; Iomab-ACT); accrual is ongoing. Pts are eligible for leukapheresis if they are ≥18 years-old w/ R/R DLBCL (de novo or transformed) following ≥2 chemoimmunotherapy regimens w/ ≥1 FDG-avid measurable lesion or B-ALL following ≥1 line of multi-agent chemotherapy (R/R following induction/consolidation; prior 2 nd/3 rd gen TKI required for pts w/ Ph+ ALL) w/ ≥5% BM involvement and/or FDG-avid extramedullary disease, ECOG performance status 0-2, and w/ appropriate organ function. Active or prior CNS disease is not exclusionary. Pts previously treated w/ CD19-targeted CAR T-cell therapy are eligible as long as CD19 expression is retained. See Fig 1B/C: Post-leukapheresis, 19-28z CAR T-cells are manufactured as previously described (Park et al., NEJM, 2018). Bridging therapy is permitted at investigator discretion. Thyroid blocking is started ≥48h pre-ARC. 131-I apamistamab 75 mCi is administered 5-7 days pre-CAR T-cell infusion to achieve total absorbed marrow dose ~200 cGy w/ remaining absorbed dose The primary objective is to determine safety/tolerability of 131-I apamistamab 75 mCi given prior to 19-28z CAR T-cells in pts w/ R/R B-ALL/DLBCL. Secondary objectives include determining incidence/severity of ICANS and CRS, anti-tumor efficacy, and 19-28z CAR T-cell expansion/persistence. Key exploratory objectives include describing the cellular microenvironment following ARC and 19-28z CAR T-cell infusion using spectral cytometry, as well as cytokine levels in peripheral blood and CRS. The trial utilizes a 3+3 design in a single cohort. If dose-limiting toxicity (severe infusion-related reactions, treatment-resistant severe CRS/ICANS, persistent regimen-related cytopenias, among others defined in protocol) is seen in 0-1 of the first 3 pts treated, then up to 6 total (up to 3 additional) pts will be treated. We have designed this study to provide preliminary data to support further investigation of CD45-targeted ARCs prior to adoptive cellular therapy. Figure 1 Figure 1. Disclosures Geyer: Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Actinium Pharmaceuticals, Inc: Research Funding; Amgen: Research Funding. Geoghegan: Actinium Pharmaceuticals, Inc: Current Employment. Reddy: Actinium Pharmaceuticals: Current Employment, Current holder of stock options in a privately-held company. Berger: Actinium Pharmaceuticals, Inc: Current Employment. Ludwig: Actinium Pharmaceuticals, Inc: Current Employment. Pandit-Taskar: Bristol Myers Squibb: Research Funding; Bayer: Research Funding; Clarity Pharma: Research Funding; Illumina: Consultancy, Honoraria; ImaginAb: Consultancy, Honoraria, Research Funding; Ymabs: Research Funding; Progenics: Consultancy, Honoraria; Medimmune/Astrazeneca: Consultancy, Honoraria; Actinium Pharmaceuticals, Inc: Consultancy, Honoraria; Janssen: Research Funding; Regeneron: Research Funding. Sauter: Genmab: Consultancy; Celgene: Consultancy, Research Funding; Precision Biosciences: Consultancy; Kite/Gilead: Consultancy; Bristol-Myers Squibb: Research Funding; GSK: Consultancy; Gamida Cell: Consultancy; Novartis: Consultancy; Spectrum Pharmaceuticals: Consultancy; Juno Therapeutics: Consultancy, Research Funding; Sanofi-Genzyme: Consultancy, Research Funding. OffLabel Disclosure: 131-I apamistamab and 19-28z CAR T-cells are investigational agents in treatment of ALL and DLBCL
- Published
- 2021
- Full Text
- View/download PDF
31. Clinical Benefit of Crenolanib, with or without Salvage Chemotherapy, in Multiply Relapsed, FLT3 Mutant AML Patients after Prior Treatment with Gilteritinib
- Author
-
Michael R. Grunwald, Boo Messahel, Aaron D Goldberg, Mark B. Geyer, Yijia Wang, Eros Di Bona, Asif Pathan, Timothy S. Pardee, Rupali Bhave, Giovanni Marconi, and Jonathan Kell
- Subjects
Prior treatment ,medicine.medical_specialty ,business.industry ,Immunology ,Salvage treatment ,Gilteritinib ,Salvage therapy ,Cell Biology ,Hematology ,Biochemistry ,Clinical trial ,chemistry.chemical_compound ,chemistry ,Tolerability ,Informed consent ,Internal medicine ,medicine ,business ,Crenolanib - Abstract
Background: Gilteritinib monotherapy provides a CR/CRh rate of 34% and a median event-free survival of 2.3 months in relapsed FLT3 mutant AML. Many AML patients (pts) who relapse after gilteritinib still express mutant FLT3 receptor and could potentially respond to FLT3 inhibition. Crenolanib is a type I, pan-FLT3 inhibitor which can be given as monotherapy or combined at full doses with standard salvage chemotherapy. Methods: We here report our experience with 7 consecutive pts who received crenolanib on compassionate basis after progressing on gilteritinib. IRB/local ethics approval was obtained prior to treatment for each pt and all pts signed informed consent forms. Results: Prior Treatment history: From Sep 2019 to July 2020, 7 consecutive pts with multiply relapsed FLT3-mutant AML who received compassionate use crenolanib after progressing on gilteritinib were identified. Four pts had relapsed after allogeneic HSCT (one pt had undergone two HSCT) and the other 3 were primary refractory despite multiple prior lines of therapy (range 2-5, median 4). Clinical presentation: At the time of treatment all pts had bone-marrow infiltration or greater than 20% circulating blasts. 1 pt did not undergo BM assessment prior to salvage chemotherapy and crenolanib but had 87% circulating blasts in the peripheral blood. One pt had cranial nerve palsy due to leukemic meningitis, another pt had extramedullary relapse in spleen and lymph nodes. Five pts had FLT3-ITD (1 of whom also had the gate-keeper FLT3-F691L mutations); 1 had FLT3-D835 and 1 had both FLT3-ITD and TKD. Crenolanib treatment: Crenolanib at 100mg TID was administered with intensive salvage therapy in 3 pts (FLAG-IDA in 2, HiDAC in 1). In the other 4 pts crenolanib was given with a palliative intent (with decitabine in 2, with azacytidine in 1 and as monotherapy in 1). Tolerability of crenolanib: Crenolanib has been clinically well tolerated, with mild nausea and vomiting reported in 3 pts which was controlled by antiemetics without causing any drug interruptions. Two pts had elevated transaminases (which resolved as concomitant medications were discontinued). There were no cardiac toxicities, pericardial effusion, fluid retention or weight gain. Response to crenolanib: At the time of this abstract, 5 of the 7 pts remain on crenolanib from a period of 18-160 days. Six of 7 pts reported clinical benefit with crenolanib. Of the 3 pts being treated with curative intent, the pt with extramedullary disease in the spleen and lymph nodes had complete resolution of her AML by PET scan. She also had exhibited clearance of bone-marrow blasts with full count recovery and became FLT3 negative. The pt with FLT3-F691 mutation and cranial nerve palsy due to CNS leukemia had clearance of CNS blasts and improvement in cranial nerve function after HIDAC, crenolanib, and intrathecal cytarabine; this patient achieved morphologic leukemia free state and FLT3 negativity in the BM. A 22-year-old pt treated with FLAG-IDA has only received 18 days of crenolanib at time of data cutoff. All pts treated with curative intent continue on crenolanib. Of the 4 pts treated with palliative intent, the pt with mutant TP53 had no benefit following crenolanib and azacytidine and died within two months. One patient, who had had two prior HSCTs, had 90% reduction in circulating blasts with azacytidine + crenolanib, but chose to discontinue treatment after 9 days. The other 2 pts continue on crenolanib 2 and 5 months after starting treatment. Conclusion: With the widespread availability of gilteritinib, more pts are now in need of treatment options after gilteritinib refractoriness or treatment failure. These pts often have resistant clones due to either acquisition of the gate-keeper FLT3-F691 mutation or RAS mutant clones. In this consecutive case series of 7 post-gilteritinib pts, treated in US and Europe, crenolanib can be given at full doses in combination with intensive salvage chemotherapy or decitabine/azacitidine. The individual-patient-level experience suggests crenolanib could effectively clear FLT3-ITD and variant FLT3-TKD mutations in pts progressing from multiple lines of treatment including gilteritinib. Clinical trials combining crenolanib with salvage chemotherapy in relapsed and refractory AML with FLT3 mutations are ongoing. More information about crenolanib compassionate use program is available by emailing compassionate@arogpharma.com. Disclosures Goldberg: Celgene: Consultancy; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Aptose: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; ADC Therapeutics: Research Funding; Aprea: Research Funding; AROG: Research Funding; Celularity: Research Funding; Dava Oncology: Honoraria; Pfizer: Research Funding. Geyer:Amgen: Research Funding. Pardee:Rafael Pharmaceuticals: Consultancy; AbbVie: Consultancy; Genentech, Inc.: Consultancy; BMS: Consultancy, Honoraria, Speakers Bureau; Karyopharm: Research Funding; Rafael: Research Funding; Celgene: Consultancy, Honoraria, Speakers Bureau; Amgen: Honoraria, Speakers Bureau; Pharmacyclics: Speakers Bureau. Grunwald:Cardinal Health: Consultancy; Amgen: Consultancy; Amgen: Consultancy; Agios: Consultancy; Merck: Consultancy; Merck: Consultancy; Abbvie: Consultancy; Agios: Consultancy; Daiichi Sankyo: Consultancy; Agios: Consultancy; Abbvie: Consultancy; Abbvie: Consultancy; Trovagene: Consultancy; Daiichi Sankyo: Consultancy; Incyte: Consultancy, Research Funding; Celgene: Consultancy; Incyte: Consultancy, Research Funding; Astellas: Consultancy; Astellas: Consultancy; Astellas: Consultancy; Genentech/Roche: Research Funding; Premier: Consultancy; Premier: Consultancy; Janssen: Research Funding; Genentech/Roche: Research Funding; Genentech/Roche: Research Funding; Forma Therapeutics: Research Funding; Merck: Research Funding; Janssen: Research Funding; Forma Therapeutics: Research Funding; Forma Therapeutics: Research Funding; Trovagene: Consultancy; Premier: Consultancy; Merck: Consultancy; Daiichi Sankyo: Consultancy; Trovagene: Consultancy; Incyte: Consultancy, Research Funding; Pfizer: Consultancy; Celgene: Consultancy; Celgene: Consultancy; Cardinal Health: Consultancy; Pfizer: Consultancy; Amgen: Consultancy; Pfizer: Consultancy; Cardinal Health: Consultancy. Wang:Arog Pharmaceuticals: Current Employment. Pathan:Arog Pharmaceuticals: Current Employment. Messahel:AROG Pharmaceuticals: Current Employment.
- Published
- 2020
- Full Text
- View/download PDF
32. BCMA-targeted CAR T-cell therapy plus radiation therapy for the treatment of refractory myeloma reveals potential synergy
- Author
-
Brigitte Senechal, Eric L. Smith, Sham Mailankody, Mark B. Geyer, Andreas Rimner, Terence J. Purdon, Isabelle Riviere, Ola Landgren, Anthony F. Daniyan, Jonathan Landa, Mette Staehr, Bianca Santomasso, Xiuyan Wang, Elena Mead, Aaron D Goldberg, and Renier J. Brentjens
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Spinal cord compression ,Internal medicine ,medicine ,Combined Modality Therapy ,Neoplasm ,Humans ,B-Cell Maturation Antigen ,Radiotherapy ,business.industry ,T-cell receptor ,Remission Induction ,Immunotherapy ,Middle Aged ,medicine.disease ,Chimeric antigen receptor ,Radiation therapy ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Female ,business ,Multiple Myeloma ,human activities - Abstract
We present a case of a patient with multiply relapsed, refractory myeloma whose clinical course showed evidence of a synergistic abscopal-like response to chimeric antigen receptor (CAR) T-cell therapy and localized radiotherapy (XRT). Shortly after receiving B-cell maturation antigen (BCMA)–targeted CAR T-cell therapy, the patient required urgent high-dose steroids and XRT for spinal cord compression. Despite the steroids, the patient had a durable systemic response that could not be attributed to XRT alone. Post-XRT findings included a second wave of fever and increased CRP and IL6, beginning 21 days after CAR T cells, which is late for cytokine-release syndrome from CAR T-cell therapy alone on this trial. Given this response, which resembled cytokine-release syndrome, immediately following XRT, we investigated changes in the patient's T-cell receptor (TCR) repertoire over 10 serial time points. Comparing T-cell diversity via Morisita's overlap indices (CD), we discovered that, although the diversity was initially stable after CAR T-cell therapy compared with baseline (CD = 0.89–0.97, baseline vs. 4 time points after CAR T cells), T-cell diversity changed after the conclusion of XRT, with >30% newly expanded TCRs (CD = 0.56–0.69, baseline vs. 4 time points after XRT). These findings suggest potential synergy between radiation and CAR T-cell therapies resulting in an abscopal-like response.
- Published
- 2019
33. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL
- Author
-
Brigitte Senechal, Meier Hsu, Elizabeth Halton, Jae H. Park, M. Lia Palomba, Isabelle Riviere, Terence J. Purdon, Yongzeng Wang, Dayenne G. van Leeuwen, Renier J. Brentjens, Xiuyan Wang, Mark B. Geyer, Michel Sadelain, Sean M. Devlin, and Yvette Bernal
- Subjects
0301 basic medicine ,Oncology ,Male ,Transplantation Conditioning ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,Phases of clinical research ,Immunotherapy, Adoptive ,chemistry.chemical_compound ,0302 clinical medicine ,Piperidines ,immune system diseases ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Receptors, Chimeric Antigen ,General Medicine ,Middle Aged ,Neoadjuvant Therapy ,Cytokine release syndrome ,Tolerability ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Ibrutinib ,Female ,Cytokine Release Syndrome ,Adult ,medicine.medical_specialty ,Lymphoma, B-Cell ,Antigens, CD19 ,Transplantation, Autologous ,Disease-Free Survival ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Aged ,Chemotherapy ,business.industry ,Adenine ,Leukapheresis ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Lymphoma ,030104 developmental biology ,Pyrimidines ,chemistry ,Drug Resistance, Neoplasm ,Pyrazoles ,Neoplasm Recurrence, Local ,Clinical Medicine ,business ,Follow-Up Studies - Abstract
BACKGROUND: Subgroups of patients with relapsed or refractory (R/R) chronic lymphocytic leukemia (CLL) exhibit suboptimal outcomes after standard therapies, including oral kinase inhibitors. We and others have previously reported on the safety and efficacy of autologous CD19-targeted CAR T cells for these patients. Here, we report safety and long-term follow-up of CAR T cell therapy with or without conditioning chemotherapy for patients with R/R CLL and indolent B cell non-Hodgkin lymphoma (B-NHL). METHODS: We conducted a phase I clinical trial investigating CD19-targeted CAR T cells incorporating a CD28 costimulatory domain (19–28z). Seventeen of twenty patients received conditioning chemotherapy prior to CAR T cell infusion. Five patients with CLL received ibrutinib at the time of autologous T cell collection and/or CAR T cell administration. RESULTS: This analysis included 16 patients with R/R CLL and 4 patients with R/R indolent B-NHL. Cytokine release syndrome (CRS) was observed in all 20 patients, but grade 3 and 4 CRS and neurological events were uncommon (10% for each). Ex vivo expansion of T cells and proportions of CAR T cells with the CD62L(+)CD127(+) immunophenotype were significantly greater (P = 0.047; CD8 subset, P = 0.0061, CD4 subset) in patients on ibrutinib at leukapheresis. Three of twelve evaluable CLL patients receiving conditioning chemotherapy achieved complete response (CR) (2 had minimal residual disease–negative CR). All patients achieving CR remained progression free at median follow-up of 53 months. CONCLUSION: Conditioning chemotherapy and 19–28z CAR T cells were acceptably tolerated across investigated dose levels in heavily pretreated patients with R/R CLL and indolent B-NHL, and a subgroup of patients achieved durable CR. Ibrutinib therapy may modulate autologous T cell phenotype. TRIAL REGISTRATION: ClinicalTrials.gov NCT00466531. FUNDING: Juno Therapeutics and NIH/National Cancer Institute Cancer Center Support Grant (P30-CA08748).
- Published
- 2019
34. Blinatumomab administered concurrently with oral tyrosine kinase inhibitor therapy is a well-tolerated consolidation strategy and eradicates measurable residual disease in adults with Philadelphia chromosome positive acute lymphoblastic leukemia
- Author
-
Martin S. Tallman, Mark B. Geyer, Amber C. King, Jae H. Park, and Jeremy J. Pappacena
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Neoplasm, Residual ,medicine.medical_treatment ,Administration, Oral ,Biochemistry ,Tyrosine-kinase inhibitor ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Antibodies, Bispecific ,Antineoplastic Combined Chemotherapy Protocols ,Philadelphia Chromosome ,Ponatinib ,Hematology ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Protein-Tyrosine Kinases ,Chemotherapy regimen ,Dasatinib ,Treatment Outcome ,030220 oncology & carcinogenesis ,Blinatumomab ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,medicine.drug_class ,Immunology ,Article ,03 medical and health sciences ,Acute lymphocytic leukemia ,Internal medicine ,medicine ,Humans ,Protein Kinase Inhibitors ,Aged ,Retrospective Studies ,Chemotherapy ,business.industry ,Imatinib ,Cell Biology ,medicine.disease ,Minimal residual disease ,Transplantation ,Consolidation Chemotherapy ,030104 developmental biology ,chemistry ,Nilotinib ,business ,030215 immunology - Abstract
Introduction: Incorporation of ABL-targeted oral tyrosine kinase inhibitors (TKIs) to conventional chemotherapy regimens has improved outcomes for adult patients (pts) with (w/) Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL). However, the addition of chemotherapy to TKIs may increase rates of infectious complications, organ toxicity, hospitalization, and mortality. As a result, we and others have investigated novel chemotherapy-sparing approaches for pts w/ Ph+ ALL. The CD3/CD19-targeted bispecific T-cell engager blinatumomab (BLIN) has demonstrated single-agent activity in pts w/ B-ALL w/ minimal residual disease (MRD) or relapsed/refractory (R/R) ALL, including R/R Ph+ ALL (Martinelli et al., J Clin Oncol, 2017). In an effort to deepen responses, we have used BLIN concomitantly w/ commercially available TKIs as consolidation and re-induction therapy. Reported experience w/ BLIN+TKI is limited. Herein we describe the observed safety and efficacy of BLIN+TKI in pts w/ Ph+ ALL w/ varying disease burden at our institution. Methods: We reviewed electronic medical records of pts ≥ 18 years (yrs) old w/ previously treated Ph+ ALL receiving BLIN w/ concomitant, FDA-approved TKIs at Memorial Sloan Kettering Cancer Center (MSKCC) who began BLIN+TKI between March 2017 and April 2018. The primary objectives were to characterize the safety/toxicity profile and rates of MRD negativity by flow cytometry (FACS) and/or quantitative real-time PCR for BCR-ABL1 transcripts following consolidation (n=9) or re-induction (n=2) w/ BLIN+TKI. Results: Eleven pts (6F, 5M) were identified (Table 1). Median age at start of BLIN+TKI was 61.2 yrs (range, 27.7-76.9). Three pts had undergone prior allogeneic hematopoietic cell transplantation (alloHCT). No pt had baseline hepatic dysfunction, central nervous system (CNS) or extramedullary (EM) disease prior to BLIN+TKI. All pts had documented CD19 expression on blasts prior to treatment. Ponatinib (PON) was the most commonly used TKI (n=7) followed by dasatinib (DAS, n=3), and nilotinib (NIL, n=1); one pt taking BLIN+PON briefly received BLIN+DAS during cycle 1. All pts were admitted for initiation of BLIN+TKI. BLIN was started at 9 mcg/day IVCI w/ escalation to 28 mcg/day IVCI on day 8 (per recommended dosing for pts w/ R/R ALL) in 10 pts and at 28 mcg/day IVCI in one pt (per flat dosing schedule described by Gökbuget et al., Blood, 2018). All 7 pts w/ MRD by FACS (n=5) and/or BCR-ABL1 PCR (n=7) prior to BLIN+TKI exhibited MRD negativity by FACS following 1 cycle of BLIN+TKI; 6 of these 7 pts achieved complete molecular response (CMR) by PCR following 1 cycle (n=5) or 2 cycles (n=1) of BLIN+TKI. All 6 remain in ongoing CMR at 1.8-15.1 months after initial achievement of CMR. Neither of the 2 pts w/ morphologic disease responded to BLIN+TKI and ultimately succumbed to their disease. The 2 pts who began BLIN+TKI in CMR have both maintained continuous CMR. Three pts have proceeded to 1st (n=2) or 2nd (n=1) alloHCT post-BLIN+TKI, without documented, relevant toxicities attributable to prior blinatumomab exposure. Median event-free and overall survival were not reached in this small group of pts, w/ median follow-up of 7.7 months among survivors (range, 3.2-16.0 months). Three pts developed grade 1 cytokine release syndrome (CRS). Notably, 2 of these pts had morphologic ALL at time of BLIN+TKI; none of these events warranted interruption of therapy or corticosteroids. No pts developed neurologic toxicity. Five pts exhibited transient transaminitis during BLIN+TKI; none reached recommended parameters to discontinue BLIN. Of these 5 pts, 4 were on concurrent PON. One pt required dose attenuation of PON; hepatic enzymes otherwise normalized w/o intervention. Conclusions: Our small series suggests that BLIN+TKI may be a safe and effective consolidation strategy for pts w/ MRD+ Ph+ ALL to achieve or sustain CMR, creating a platform for alloHCT or other post-remission therapy. Observed rates of CRS were higher and neurologic toxicity appeared lower (none documented) than in other studies of BLIN in pts w/ MRD (Gökbuget et al., Blood, 2018). Higher risk of transaminitis was noted in pts receiving BLIN+PON and warrants further observation, particularly as 28 mcg/day IVCI flat dosing is increasingly used for pts w/ MRD. Earlier incorporation of BLIN+TKI into treatment paradigms for Ph+ ALL may limit toxicity while deepening response and maintaining CMR. Disclosures King: Genentech: Other: Advisory Board . Tallman:Daiichi-Sankyo: Other: Advisory board; Orsenix: Other: Advisory board; Cellerant: Research Funding; ADC Therapeutics: Research Funding; AbbVie: Research Funding; AROG: Research Funding; BioSight: Other: Advisory board. Park:Pfizer: Consultancy; Juno Therapeutics: Consultancy, Research Funding; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Consultancy; Novartis: Consultancy; Kite Pharma: Consultancy; Adaptive Biotechnologies: Consultancy; Shire: Consultancy. Geyer:Dava Oncology: Honoraria.
- Published
- 2019
35. Inotuzumab ozogamicin as chemotherapy-sparing salvage in a 67-year-old man with primary refractory B-cell acute lymphoblastic leukemia with high-risk genomic features
- Author
-
Mark B. Geyer, Filiz Sen, and Xiaochuan Yang
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Disease ,Acute lymphoblastic leukemia ,Philadelphia chromosome ,lcsh:RC254-282 ,Article ,Inotuzumab ozogamicin ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Chemotherapy ,business.industry ,Induction chemotherapy ,Hematology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Minimal residual disease ,Respiratory failure ,Older adults ,030220 oncology & carcinogenesis ,Immunotherapy ,business ,030215 immunology ,medicine.drug - Abstract
Older adults with acute lymphoblastic leukemia (ALL) continue to have a poor prognosis, in part due to greater chemotherapy-related toxicities. We herein report a 67-year-old man with Philadelphia chromosome (Ph)-negative B-cell ALL, who exhibited refractoriness to 3 different regimens of induction chemotherapy and experienced multiple complications including intracranial bleeding and respiratory failure, who achieved minimal residual disease (MRD)-negative complete response (CR) after a single cycle of inotuzumab ozogamicin (IO). His ALL was characterized by several high-risk mutations, which may have contributed to chemotherapy-refractory disease. Our case supports incorporating IO into front-line induction regimens for older adults with high-risk B-cell ALL. Keywords: Acute lymphoblastic leukemia, Inotuzumab ozogamicin, Older adults, Immunotherapy
- Published
- 2019
36. Review: Current clinical applications of chimeric antigen receptor (CAR) modified T cells
- Author
-
Mark B. Geyer and Renier J. Brentjens
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,hemic and lymphatic diseases ,Internal medicine ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Genetics (clinical) ,Multiple myeloma ,Clinical Trials as Topic ,Transplantation ,Tumor microenvironment ,business.industry ,Cell Biology ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,Clinical trial ,Cytokine release syndrome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Refractory Chronic Lymphocytic Leukemia ,business - Abstract
The past several years have been marked by extraordinary advances in clinical applications of immunotherapy. In particular, adoptive cellular therapy utilizing chimeric antigen receptor (CAR) modified T cells targeted to CD19 has demonstrated substantial clinical efficacy in children and adults with relapsed or refractory B cell acute lymphoblastic leukemia (B-ALL), and durable clinical benefit in a smaller subset of patients with relapsed or refractory chronic lymphocytic leukemia (CLL) or B cell non-Hodgkin lymphoma (B-NHL). Early phase clinical trials are presently assessing CAR T cell safety and efficacy in additional malignancies. Herein, we discuss clinical results from the largest series to date investigating CD19-targeted CAR T cells in B-ALL, CLL, and B-NHL, including discussion of differences in CAR T cell design and production and treatment approach, as well as clinical efficacy, nature of severe cytokine release syndrome and neurologic toxicities, and CAR T cell expansion and persistence. We additionally review the current and forthcoming use of CAR T cells in multiple myeloma and several solid tumors, and highlight challenges and opportunities afforded by the current state of CAR T cell therapies, including strategies to overcome inhibitory aspects of the tumor microenvironment and enhance antitumor efficacy.
- Published
- 2016
- Full Text
- View/download PDF
37. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date
- Author
-
Jae H. Park, Renier J. Brentjens, and Mark B. Geyer
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Adoptive cell transfer ,T-Lymphocytes ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Pharmacology ,Biochemistry ,CD19 ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Internal medicine ,medicine ,Animals ,Humans ,In patient ,B-Lymphocytes ,biology ,business.industry ,Review Series ,Cell Biology ,Hematology ,Adoptive Transfer ,Chimeric antigen receptor ,Clinical trial ,030104 developmental biology ,Hematologic Neoplasms ,030220 oncology & carcinogenesis ,biology.protein ,Car t cells ,business ,human activities - Abstract
Adoptive transfer of T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 has produced impressive results in treating patients with B-cell malignancies. Although these CAR-modified T cells target the same antigen, the designs of CARs vary as well as several key aspects of the clinical trials in which these CARs have been studied. It is unclear whether these differences have any impact on clinical outcome and treatment-related toxicities. Herein, we review clinical results reflecting the investigational use of CD19-targeted CAR T-cell therapeutics in patients with B-cell hematologic malignancies, in light of differences in CAR design and production, and outline the limitations inherent in comparing outcomes between studies.
- Published
- 2016
- Full Text
- View/download PDF
38. Phase II Study of Blinatumomab and Concurrent Oral Tyrosine Kinase Inhibitor Therapy As Consolidation and Maintenance Therapy for Patients with Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia Following Chemotherapy-Sparing Induction
- Author
-
Mark B. Geyer, Amber C. King, Justin C. O'Brien, and Jae H. Park
- Subjects
Oncology ,Chemotherapy ,medicine.medical_specialty ,education.field_of_study ,business.industry ,medicine.medical_treatment ,Immunology ,Population ,Phases of clinical research ,Cell Biology ,Hematology ,Biochemistry ,Minimal residual disease ,Dasatinib ,Maintenance therapy ,Prednisone ,Internal medicine ,medicine ,Blinatumomab ,business ,education ,medicine.drug - Abstract
Background: Combining oral ABL-targeted tyrosine kinase inhibitors (TKIs) with (w/) multi-agent chemotherapy has improved the long-term disease-free survival of adults w/ Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL). However, Ph+ ALL occurs more commonly in older adults, and toxicities of multi-agent chemotherapy, including sequelae of prolonged myelosuppression, are amplified in this population. Others have previously reported rates of morphologic complete response (mCR) approaching 100% among adults w/ Ph+ ALL treated w/ corticosteroids (CS) and dasatinib (DAS) alone as induction therapy, but w/ low rates of minimal residual disease (MRD) negativity by flow cytometry (FACS) and BCR-ABL1 PCR (complete molecular response, CMR) and high rates of relapse in the absence of further consolidation. The bispecific T-cell engager blinatumomab (BLIN) has considerable efficacy in clearing MRD in patients (pts) w/ Ph- B-cell ALL. We have previously reported our institutional experience combining ABL TKIs w/ BLIN in pts w/ Ph+ ALL and MRD, including encouraging safety data and high rates of MRD eradication (King/Geyer et al., Leuk Res, 2019). The ongoing D-ALBA study (GIMEMA LAL2116) is also investigating BLIN + DAS consolidation following 12 weeks of induction (prednisone + DAS followed by DAS), w/out protocol-specified maintenance, and has demonstrated preliminary evidence of efficacy (Chiaretti et al., ASH Meeting, 2019). As such, we designed a phase II study of BLIN as part of a chemotherapy sparing strategy in pts w/ Ph+ ALL (BLISSPHALL), introducing BLIN as early as 6 weeks into treatment for pts in morphologic CR, w/ aim of enhancing early MRD negativity and suppressing resistant clones early in disease course. Study design and methods: Our institution is leading a phase II trial of TKI + BLIN consolidation and maintenance in adults w/ newly-diagnosed Ph+ ALL, w/ potential multicenter expansion (NCT04329325). Pts are eligible if they are ≥18 years-old w/ Ph+ ALL confirmed by cytogenetic or molecular studies, ECOG performance status 0-2, w/out prior therapy for ALL beyond CS, hydroxyurea, or intrathecal chemotherapy, w/out known active extramedullary disease and/or CNS-3 disease, and w/ appropriate organ function. See Figure 1: pts will receive a CS pre-phase (days [d] -6 - 0) followed by modified GIMEMA LAL1205 induction (dexamethasone [DEX] 10 mg/m2 [max 24 mg/d], d1-24, tapered off d25-32) + DAS 140 mg/d (dose adjustments or TKI change permitted per protocol) w/ intrathecal methotrexate (IT MTX) d22, 43 and bone marrow (BM) assessments including FACS and BCR-ABL1 PCR. Pts in mCR on d43 (or optional reassessment ≤ 3 weeks later) will be eligible to proceed to consolidation w/ 3 cycles (C) BLIN 28 mcg/d IVCI, d1-28, concurrent w/ TKI, w/ 14d off BLIN between cycles and BM MRD assessment/IT MTX after each cycle. C1 BLIN + TKI is required to begin inpatient x72 hours. TKI is given continuously including between BLIN cycles. Pts in CMR after consolidation may proceed to maintenance (C4-7 BLIN + TKI, 28d off between cycles). Pts may come off study to proceed to hematopoietic cell transplant (HCT) at any point, though it is recommended such pts receive ≥2C of BLIN + TKI. The primary objective is to determine the proportion of evaluable pts achieving CMR by the end of consolidation (≤ 3C BLIN + TKI). Secondary objectives include safety/toxicity of BLIN + DAS, duration of CMR, incidence of relapse, event-free/overall survival. Exploratory objectives include safety/toxicity of non-DAS TKIs + BLIN, defining patterns/mechanisms of resistance to BLIN+TKI (including ABL kinase mutations), and outcomes among pts not undergoing HCT. The trial utilizes a Simon's minimax two-stage design; 20% CMR rate is considered not promising, a 50% CMR rate is considered promising, and probabilities of type I/II error are set at 0.10/0.10. If ≥ 3 of the first 10 pts achieve CMR we will continue enrollment to max 17 pts. If ≥ 6 pts achieve CMR, then BLIN + TKI will be considered promising for further investigation. The investigators are hopeful this study will add to the currently limited prospective data supporting TKI + BLIN consolidation/maintenance for pts w/ Ph+ ALL and efforts to develop chemotherapy-sparing and immunotherapeutic strategies for older pts w/ ALL. Disclosures Geyer: Amgen: Research Funding. King:Abbvie: Other: advisory board. Park:Novartis: Consultancy; Genentech/Roche: Research Funding; Intellia: Consultancy; Artiva: Membership on an entity's Board of Directors or advisory committees; Fate Therapeutics: Research Funding; Takeda: Consultancy, Research Funding; Servier: Consultancy, Research Funding; AstraZeneca: Consultancy; Allogene: Consultancy; Incyte: Consultancy, Research Funding; Kite: Consultancy, Research Funding; Juno Therapeutics: Research Funding; GSK: Consultancy; Autolus: Consultancy, Research Funding; Minverva: Consultancy; Amgen: Consultancy, Research Funding. OffLabel Disclosure: Blinatumomab is not approved to be given in combination with ABL tyrosine kinase inhibitors for treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia.
- Published
- 2020
- Full Text
- View/download PDF
39. Abstract 4248: Multispectral immunofluorescence identifies pS6 as a biomarker of intrinsic resistance to ruxolitinib in patients with relapsed/refractory T-cell lymphomas
- Author
-
Lauren Pomerantz, Steven M. Horwitz, Eric N. Jacobsen, David M. Weinstock, Ahmet Dogan, Nivetha Ganesan, Obadi Obadi, Johnathan Schatz, Ariela Noy, David J. Straus, Allison Sigler, Theresa Davey, Helen Hancock, Andrea Knezevic, Giorgio Inghirami, Venkatraman E. Seshan, Mark B. Geyer, Carlissa Onwasigwe, Alison J. Moskowitz, Christine Jarjies, Paola Ghione, Sarah J. Noor, Jia Ruan, Travis J. Hollmann, Patricia L. Myskowski, Priadarshini Kumar, and Natasha Galasso
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Ruxolitinib ,medicine.diagnostic_test ,business.industry ,Bortezomib ,T cell ,Cancer ,medicine.disease ,Duvelisib ,Romidepsin ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Internal medicine ,Biopsy ,medicine ,business ,CD8 ,medicine.drug - Abstract
Introduction: Peripheral and cutaneous T cell lymphomas (TCL) are highly heterogeneous diseases that in the relapsed/refractory (R/R) stage have poor outcome. JAK/STAT, Pi3K/AKT and MAP kinase signaling are commonly involved in the pathogenesis of these lymphomas, and the efficacy of small molecule inhibitors targeting these pathways is being assessed in phase II trials. However, biomarkers to define patients most likely to benefit from specific inhibitors are lacking and mechanisms of acquired resistance in TCLs have not been reported. Methods: We are conducting two early phase trials in R/R TCL: 1) NCT02974647 targets JAK1/JAK2 with ruxolitinib (RUX, a JAK 1-2 inhibitor) and has enrolled 53 patients and 2) NCT02783625 targets Pi3K/AKT with duvelisib (DUV, a Pi3K gamma-delta inhibitor) in combination with either romidepsin or bortezomib and has enrolled 92 patients. We analyzed pre-, on and post- treatment biopsies with multiplex immunofluorescence (mIF) using the Vectra platform, and analyzed images for marker expression and cellular location using HALO software. We designed multiple mIF panels with 6 markers per slide, 3 identifying TCL cells based on disease-specific phenotypes (e.g. CD4+PD1+CD8- for angioimmunoblastic TCL), pSTAT3,5, pS6, and a macrophage marker (CD68). Areas of TCL involvement within biopsy slides were manually defined during image analysis based on mIF and comparison to clinical immunohistochemistry. Differences in marker expressions were analyzed with the Wilcoxon test using the program R. Results: We analyzed 53 biopsies from 39 patients with high-quality mIF images, of which 17 were from lymph nodes and 36 were from extranodal sites. Of 39 patients, 13 were treated with RUX (9 biopsies pre-, 7 on, 4 post- treatment) and 26 with DUV (20 biopsies pre-, 6 on, 10 post- treatment). Median number of total TCL cells in pre-treatment biopsies was 1041 (range, 97-6463) and median TCL cell fraction within involved areas was 27.86% (range, 5.12-88.02%). Median macrophage fraction within involved areas was 5.92% (range, 2.89-11.88%). For either agent, response to treatment was not associated with: 1) quantity of macrophages (p=0.1 for responders vs non-responders to DUV, p=0.53 for RUX), 2) average distance between TCL cells and closest macrophage, 3) TCL cell fraction within involved areas, or 4) fraction of TCL cells expressing pSTAT3/5 (Figure). In contrast, pS6 expression in Conclusions: mIF is a feasible platform for biomarker discovery and translation in TCL, a set of lymphomas with heterogeneous immunophenotypes. pS6 expression in Citation Format: Paola Ghione, Alison Moskowitz, Priadarshini Kumar, Andrea Knezevic, Venkatraman Seshan, Eric Jacobsen, Jia Ruan, Johnathan Schatz, Sarah Noor, Patricia Myskowski, Helen Hancock, Theresa Davey, Obadi Obadi, Carlissa Onwasigwe, Nivetha Ganesan, Lauren Pomerantz, Christine Jarjies, Allison Sigler, Mark Geyer, Ariela Noy, David Straus, Natasha Galasso, Giorgio Inghirami, Steven Horwitz, David Weinstock, Travis Hollmann, Ahmet Dogan. Multispectral immunofluorescence identifies pS6 as a biomarker of intrinsic resistance to ruxolitinib in patients with relapsed/refractory T-cell lymphomas [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4248.
- Published
- 2020
- Full Text
- View/download PDF
40. Chimeric Antigen Receptor-T Cells for Leukemias in Adults: Methods, Data and Challenges
- Author
-
Jae H. Park, Renier J. Brentjens, and Mark B. Geyer
- Subjects
Adoptive cell transfer ,business.industry ,medicine.medical_treatment ,T cell ,T-cell receptor ,CD28 ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Leukemia ,medicine.anatomical_structure ,medicine ,Cancer research ,Hairy cell leukemia ,business - Abstract
Within the past several years, renewed interest in immunotherapy has been observed in multiple fields of oncology, including antibody-based therapeutics (e.g., checkpoint blockade) and adoptive cellular therapies. In the field of adult leukemias, such interest has been driven by the limitations of presently available therapies to induce durable remissions reliably in patients with relapsed and refractory leukemia. The adoptive transfer of genetically modified autologous T-cells aims to rapidly establish specific antitumor activity. This strategy requires targeting of autologous T-cells by means of a transgene-encoded antigen receptor, consisting of a chimeric antigen receptor (CAR), as will be discussed herein, or T-cell receptor (TCR) chains. CD19 is nearly universally expressed by B-ALL, CLL, and hairy cell leukemia, while not expressed on normal tissues other than B-cells, including multipotent hematopoietic progenitor cells. Multiple generations of CARs have been developed and investigated in clinical studies and will be discussed in this review. In this chapter, we review clinical outcomes of adults with leukemia treated with CAR T-cells, toxicities associated with CAR T-cell administration, present challenges limiting therapeutic efficacy, and future directions, including novel targets and enhancements to improve antileukemic activity.
- Published
- 2018
- Full Text
- View/download PDF
41. Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia
- Author
-
Jessica Schulman, Akshar Patel, Ross L. Levine, Sergio Giralt, Sheng F. Cai, Erin McGovern, Justin Taylor, Virginia M. Klimek, Martin S. Tallman, Sinnifer Sim, Sean M. Devlin, Andrew Dunbar, Charlene C. Kabel, Mark B. Geyer, Nghia T. Nguyen, Aaron D. Viny, Yanming Zhang, Christopher Famulare, Wenbin Xiao, Kamal Menghrajani, Zachary D. Epstein-Peterson, Minal Patel, Aaron D Goldberg, Mikhail Roshal, Jacob L. Glass, and Bartlomiej Getta
- Subjects
Oncology ,Adult ,Male ,Acute Myeloid Leukemia ,medicine.medical_specialty ,Myeloid ,Neoplasm, Residual ,Plasmacytoid dendritic cell ,Article ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,Internal medicine ,Medicine ,Humans ,Plasmacytoid dendritic cell differentiation ,Aged ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Myeloid leukemia ,Hematology ,Dendritic cell ,Dendritic Cells ,Middle Aged ,medicine.disease ,Prognosis ,Combined Modality Therapy ,3. Good health ,Survival Rate ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Case-Control Studies ,Female ,Neoplasm Recurrence, Local ,business ,030215 immunology ,Follow-Up Studies - Abstract
Measurable residual disease is associated with inferior outcomes in patients with acute myeloid leukemia (AML). Measurable residual disease monitoring enhances risk stratification and may guide therapeutic intervention. The European LeukemiaNet working party recently came to a consensus recommendation incorporating leukemia associated immunophenotype-based different from normal approach by multi-color flow cytometry for measurable residual disease evaluation. However, the analytical approach is highly expertise-dependent and difficult to standardize. Here we demonstrate that loss of plasmacytoid dendritic cell differentiation after 7+3 induction in AML is highly specific for measurable residual disease positivity (specificity 97.4%) in a uniformly treated patient cohort. Moreover, loss of plasmacytoid dendritic cell differentiation as determined by a blast-to-plasmacytoid dendritic cell ratio >10 was strongly associated with inferior overall and relapse-free survival (RFS) [Hazard ratio 2.79, 95% confidence interval (95%CI): 0.98-7.97; P=0.077) and 3.83 (95%CI: 1.51-9.74; P=0.007), respectively), which is similar in magnitude to measurable residual disease positivity. Importantly, measurable residual disease positive patients who reconstituted plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio
- Published
- 2018
42. Autologous CD19-Targeted CAR T Cells in Patients with Residual CLL following Initial Purine Analog-Based Therapy
- Author
-
Elizabeth Halton, Brigitte Senechal, Meier Hsu, Jae H. Park, Renier J. Brentjens, Sean M. Devlin, Isabelle Riviere, Yongzeng Wang, Jürgen Rademaker, Nicole Lamanna, Xiuyan Wang, Mark B. Geyer, Michel Sadelain, and Terence J. Purdon
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,Neoplasm, Residual ,Cyclophosphamide ,Chronic lymphocytic leukemia ,T-Lymphocytes ,Antigens, CD19 ,Purine analogue ,Immunotherapy, Adoptive ,Transplantation, Autologous ,03 medical and health sciences ,0302 clinical medicine ,Chemoimmunotherapy ,Behavior Therapy ,Internal medicine ,hemic and lymphatic diseases ,Drug Discovery ,Genetics ,Medicine ,Pentostatin ,Humans ,Molecular Biology ,Aged ,Pharmacology ,business.industry ,Middle Aged ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Chimeric antigen receptor ,Cytokine release syndrome ,030104 developmental biology ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,Rituximab ,Original Article ,Female ,business ,medicine.drug - Abstract
Patients with residual chronic lymphocytic leukemia (CLL) following initial purine analog-based chemoimmunotherapy exhibit a shorter duration of response and may benefit from novel therapeutic strategies. We and others have previously described the safety and efficacy of autologous T cells modified to express anti-CD19 chimeric antigen receptors (CARs) in patients with relapsed or refractory B cell acute lymphoblastic leukemia and CLL. Here we report the use of CD19-targeted CAR T cells incorporating the intracellular signaling domain of CD28 (19-28z) as a consolidative therapy in 8 patients with residual CLL following first-line chemoimmunotherapy with pentostatin, cyclophosphamide, and rituximab. Outpatients received low-dose conditioning therapy with cyclophosphamide (600 mg/m(2)), followed by escalating doses of 3 × 10(6), 1 × 10(7), or 3 × 10(7) 19-28z CAR T cells/kg. An objective response was observed in 3 of 8 patients (38%), with a clinically complete response lasting more than 28 months observed in two patients. Self-limited fevers were observed post-CAR T cell infusion in 4 patients, contemporaneous with elevations in interleukin-6 (IL-6), IL-10, IL-2, and TGF-α. None developed severe cytokine release syndrome or neurotoxicity. CAR T cells were detectable post-infusion in 4 patients, with a longest observed persistence of 48 days by qPCR. Further strategies to enhance CAR T cell efficacy in CLL are under investigation.
- Published
- 2018
43. Thirty Day Resource Utilization after Chimeric Antigen Receptor (CAR) T Cell Infusion for Hematologic Malignancies
- Author
-
Elena Mead, Connie W. Batlevi, Miguel-Angel Perales, Anas Younes, Eric L. Smith, Bianca Santomasso, Gunjan L. Shah, M. Lia Palomba, Elaine Duck, Renier J. Brentjens, Paul Sabbatini, Peter B. Bach, Jae H. Park, Sergio Giralt, Elizabeth Halton, Mark B. Geyer, Craig S. Sauter, and Sham Mailankody
- Subjects
Transplantation ,medicine.medical_specialty ,Chemotherapy ,medicine.diagnostic_test ,business.industry ,Chronic lymphocytic leukemia ,medicine.medical_treatment ,T cell ,Hematology ,medicine.disease ,Intensive care unit ,law.invention ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,law ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Outpatient clinic ,Liver function tests ,business ,Multiple myeloma ,030215 immunology - Abstract
Background We aimed to characterize institutional resources utilized (other than T cell collection and CAR T cell manufacturing and infusion) peri-chimeric antigen receptor-modified (CAR) T cells administration. Methods Adult patients treated on selected investigator-initiated clinical trials of CAR T cell therapy at Memorial Sloan Kettering Cancer Center were identified from the institutional database. Utilization data was collected from the start of admission for CAR T cell infusion through the end of the initial admission for infusion or through 30 days following initial CAR T cell infusion, whichever was longer. Descriptive statistics were used to analyze the data. Results Between 6/2007 to 4/2018, 106 pts (56 pts (53%) B-cell acute lymphoblastic leukemia (ALL), 37 (35%) chronic lymphocytic leukemia (CLL), B-cell non-Hodgkin lymphoma (NHL), and 13 (12%) multiple myeloma (MM)) on 4 clinical trials receiving inpatient CAR T cell infusions. The median age was 53 yrs (range 22-77) with 65% male. The median length of stay for the admission during which CAR T cells was given was 23 days (range 4 -133), with ALL patients admitted longer (Figure 1). 43 (41%) spent at least one day in the intensive care unit (ICU), with a range of 0-43 days. Outpatient clinic visits through day 30 post CAR T cell infusion occurred in 57 (53%) patients. A total of 62,953 laboratory panels and 1,190 radiology studies done during the study time frame. Fourteen percent of the labs were complete blood counts, basic or comprehensive metabolic panels, or liver function tests. For the total population, ALL, CLL/NHL, MM, there were a median of 63.5 (range 13-368), 91.5 (21-368), 47 (13-167), and 51 (range 38-113) of these panels done per patient during the time frame, respectively. Among the radiology studies, 25% were CT scans, 10% MRIs, 7% PET scans, 5% ultrasounds, and 53% x-rays, with differences in patterns of use by disease type (Figure 2). Chemotherapy accounted for 328 units (0.8%) of the 41,331 units of medications given in this time frame. The median medication units per patient was 255 (range 35-2091), 423 (range 35-2091), 200 (range 41-1190), and 207 (range 90-666) for the total population, and the ALL, CLL/NHL, and MM pts, respectively. Thirty-two doses of tocilizumab were given to 25 patients (24%), with ALL pts receiving 23 of those doses (72%). Conclusion CAR T cell therapy utilizes many resources and can create challenges in institutional resource capacity. Identifying these resources will allow for better allocation and care delivery. Further refinement of CAR T cell products and improvements in CAR T cell-related toxicity management may permit safer delivery of this therapy and reduce costs per patient. Comparison with alternative therapies and analysis of resource utilization for the commercial CAR T cell products is warranted.
- Published
- 2019
- Full Text
- View/download PDF
44. The safety and efficacy of clofarabine in combination with high-dose cytarabine and total body irradiation myeloablative conditioning and allogeneic stem cell transplantation in children, adolescents, and young adults (CAYA) with poor-risk acute leukemia
- Author
-
Mitchell S. Cairo, Jessica Hochberg, M. Fevzi Ozkaynak, Lauren Harrison, Alfred P. Gillio, Alexandra Cheerva, Theodore B. Moore, Jennifer Krajewski, Julie Talano, Lee Ann Baxter-Lowe, Stacey Zahler, O. Militano, Nan Chen, Mark C. Walters, Mark B. Geyer, and Carl V. Hamby
- Subjects
Myeloid ,Oncology ,Male ,Transplantation Conditioning ,0302 clinical medicine ,Stem Cell Research - Nonembryonic - Human ,Antineoplastic Combined Chemotherapy Protocols ,Clofarabine ,Child ,Cancer ,Pediatric ,Acute leukemia ,Leukemia ,Cytarabine ,Hematopoietic Stem Cell Transplantation ,Hematology ,Total body irradiation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Leukemia, Myeloid, Acute ,Treatment Outcome ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Child, Preschool ,Acute Disease ,Female ,Whole-Body Irradiation ,medicine.drug ,Homologous ,Pediatric Research Initiative ,medicine.medical_specialty ,Platelet Engraftment ,Adolescent ,Pediatric Cancer ,Childhood Leukemia ,Clinical Trials and Supportive Activities ,Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Acute ,03 medical and health sciences ,Young Adult ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Preschool ,Transplantation ,business.industry ,Evaluation of treatments and therapeutic interventions ,Infant ,Myeloablative Agonists ,Stem Cell Research ,medicine.disease ,Orphan Drug ,Good Health and Well Being ,Relative risk ,business ,030215 immunology - Abstract
Acute leukemias in children with CR3, refractory relapse, or induction failure (IF) have a poor prognosis. Clofarabine has single agent activity in relapsed leukemia and synergy with cytarabine. We sought to determine the safety and overall survival in a Phase I/II trial of conditioning with clofarabine (doses 40 - 52 mg/m2), cytarabine 1000 mg/m2, and 1200 cGy TBI followed by alloSCT in children, adolescents, and young adults with poor-risk leukemia. Thirty-seven patients; Age 12 years (1-22 years); ALL/AML: 34:3 (18 IF, 10 CR3, 13 refractory relapse); 15 related, 22 unrelated donors. Probabilities of neutrophil, platelet engraftment, acute GvHD, and chronic GvHD were 94%, 84%, 49%, and 30%, respectively. Probability of day 100 TRM was 8.1%. 2-year EFS (event free survival) and OS (overall survival) were 38.6% (CI95: 23-54%), and 41.3% (CI95: 25-57%). Multivariate analysis demonstrated overt disease at time of transplant (relative risk (RR) 3.65, CI95: 1.35-9.89, P = 0.011) and umbilical cord blood source (RR 2.17, CI95: 1.33-4.15, P = 0.019) to be predictors of worse EFS/OS. This novel myeloablative conditioning regimen followed by alloSCT is safe and well tolerated in CAYA with very poor-risk ALL or AML. Further investigation in CAYA with better risk ALL and AML undergoing alloSCT is warranted.
- Published
- 2017
45. Concurrent therapy of chronic lymphocytic leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia utilizing CD19-targeted CAR T-cells
- Author
-
Brigitte Senechal, Xiuyan Wang, Renier J. Brentjens, Marco L. Davila, Isabelle Riviere, Mark B. Geyer, Yongzeng Wang, Shwetha H. Manjunath, Jae H. Park, Corey Cutler, Jane L. Liesveld, Michel Sadelain, and Andrew G. Evans
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,CD19 ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,law ,medicine ,Receptor ,Philadelphia Chromosome Positive ,biology ,business.industry ,Hematology ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Recombinant DNA ,business - Abstract
Chimeric antigen receptors (CARs) are recombinant receptors composed of an antibody-derived extracellular single-chain variable fragment linked to a costimulatory signaling domain combined with the...
- Published
- 2017
46. Overall survival among older US adults with ALL remains low despite modest improvement since 1980: SEER analysis
- Author
-
Mark B. Geyer, Martin S. Tallman, Jae H. Park, Sean M. Devlin, Meier Hsu, and Dan Douer
- Subjects
0301 basic medicine ,Oncology ,Gerontology ,Adult ,Male ,medicine.medical_specialty ,Disease free survival ,Databases, Factual ,Lymphoblastic Leukemia ,Immunology ,MEDLINE ,Letters to Blood ,Biochemistry ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Seer program ,medicine ,Overall survival ,Cooperative group ,Humans ,Survival rate ,Extramural ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Survival Rate ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,business ,SEER Program - Abstract
To the editor: Over the past 4 decades, outcomes have improved dramatically among pediatric patients with acute lymphoblastic leukemia (ALL), with observed cure rates now >80% in developed countries.[1][1] This progress can be attributed, in part, to large cooperative group studies, advances in
- Published
- 2017
47. Enhanced Survival During Experimental Listeria monocytogenes Sepsis in Neonatal Mice Prophylactically Treated With Th1 and Macrophage Immunoregulatory Cytokines and Mediators
- Author
-
Kavita Radhakrishnan, Mitchell S. Cairo, Janet Ayello, Mark B. Geyer, Carmella van de Ven, and Mandhir S. Suri
- Subjects
Microbiology (medical) ,medicine.disease_cause ,Chemoprevention ,Sepsis ,Immune system ,Listeria monocytogenes ,Interferon ,medicine ,Animals ,Macrophage ,business.industry ,Macrophages ,Lethal dose ,Interleukin ,Th1 Cells ,medicine.disease ,Survival Analysis ,Mice, Inbred C57BL ,Disease Models, Animal ,Treatment Outcome ,Infectious Diseases ,Animals, Newborn ,Cord blood ,Pediatrics, Perinatology and Child Health ,Immunology ,Cytokines ,Immunotherapy ,business ,medicine.drug - Abstract
Background Impairments in T-cell and macrophage-mediated host defense lead to increased infection-related morbidity and mortality in neonates, partly because of immaturity in T helper (Th)1 function. Listeria monocytogenes (Lm) is an intracellular pathogen disproportionately causing severe disease among neonates and the immunocompromised. Intact macrophage and Th1-mediated immune responses are critical for Lm clearance. We and others have previously demonstrated downregulation of Th1 and macrophage immunoregulatory cytokines in cord blood versus adult peripheral blood. We sought to determine whether therapeutic or prophylactic single agent or combination recombinant murine interleukin (rmIL)-12, rmIL-18, recombinant murine macrophage-colony stimulating factor (rmM-CSF), recombinant murine granulocyte macrophage-colony stimulating factor (rmGM-CSF) and recombinant murine interferon (rmIFN)-γ would enhance survival during experimental neonatal Lm sepsis. Methods A 90% lethal dose (LD90) of Lm was established in C57/BL/6 neonatal mice. rmIL-12, rmIL-18, rmM-CSF, rmGM-CSF and rmIFN-γ were administered singly or sequentially, before or after LD90 Lm inoculation; ampicillin was administered 24 hours after inoculation. Results Therapeutic doses of rmIL-12, rmIL-18, rmM-CSF, rmGM-CSF and rmIFN-γ as single agents and sequential therapy with rmM-CSF + (rmIL-12 and/or rmIL-18) + rmIFN-γ in addition to ampicillin were not associated with increased survival. However, prophylactic single doses of rmIL-12, rmIL-18, rmM-CSF and rmIFN-γ and prophylactic sequential doses of rmM-CSF + (rmIL-12 and/or rmIL-18) + rmIFN-γ in addition to ampicillin were associated with significantly enhanced survival compared with ampicillin alone. Conclusions These data suggest prophylactic administration of macrophage and Th1 immunoregulatory cytokines can potentially overcome deficits in neonatal immunity to protect against Lm.
- Published
- 2014
- Full Text
- View/download PDF
48. A Comparison of Bronchoalveolar Lavage versus Lung Biopsy in Pediatric Recipients after Stem Cell Transplantation
- Author
-
Phyllis Della-Letta, Miguel Muniz, William Middlesworth, E. Qualter, Michael R. Bye, Erin Morris, Zhezhen Jin, Bachir Alobeid, Lauren Harrison, Carmella van de Ven, Mitchell S. Cairo, Prakash Satwani, Kavita Radhakrishnan, Gerald Behr, Mark B. Geyer, and Angela M. Ricci
- Subjects
Lung Diseases ,Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,medicine.medical_treatment ,Lung biopsy ,Hematopoietic stem cell transplantation ,Bronchalveolar lavage ,Bronchoalveolar Lavage ,Pediatrics ,Gastroenterology ,Article ,Cohort Studies ,Autologous stem-cell transplantation ,Internal medicine ,Biopsy ,medicine ,Humans ,Transplantation, Homologous ,Child ,Transplantation ,medicine.diagnostic_test ,business.industry ,Hematopoietic Stem Cell Transplantation ,Stem cell transplantation ,Hematology ,respiratory system ,Surgery ,Bronchoalveolar lavage ,Child, Preschool ,Female ,Chest radiograph ,business - Abstract
Bronchoalveolar lavage (BAL) has been a useful initial diagnostic tool in the evaluation of pulmonary complications after hematopoietic stem cell transplantation (HSCT); however, the diagnostic sensitivity, prevalence, and outcome after BAL versus lung biopsy (LB) in pediatric HSCT patients remains to be determined. We reviewed 193 pediatric HSCT recipients who underwent a total of 235 HSCTs. Sixty-five patients (34%) underwent a total of 101 BALs for fever, respiratory distress, and/or pulmonary infiltrates on chest radiograph and/or computed tomography scan. The 1-year probability of undergoing BAL was 43.0% after allogeneic stem cell transplantation (alloSCT) and 8.5% after autologous stem cell transplantation (autoSCT) (P = .001). Sixteen of the 193 patients (8%) patients underwent 19 LBs. The probability of undergoing LB at 1 year after HSCT was 9.3%. No grade III or IV adverse events related to either procedure were observed. Of the 101 BALs performed, 40% (n = 40) were diagnostic, with a majority revealing a bacterial pathogen. Among the 19 LBs performed, 94% identified an etiology. In multivariate analysis, myeloablative conditioning alloSCT conferred the highest risk of requiring a BAL (hazard ratio [HR],8.5; P = .0002). The probability of 2-year overall survival was 20.2% in patients who underwent BAL, 17.5% for patients who underwent biopsy, and 67.4% for patients who had neither procedure. In multivariate analysis, only the requirement of a BAL was independently associated with an increased risk of mortality (HR, 2.96; P
- Published
- 2014
- Full Text
- View/download PDF
49. Reduced toxicity, myeloablative conditioning with BU, fludarabine, alemtuzumab and SCT from sibling donors in children with sickle cell disease
- Author
-
Mark B. Geyer, R. Hawks, M T Lee, Erin Morris, Joseph Schwartz, Prakash Satwani, M Licursi, Mitchell S. Cairo, Courtney Baker, G. Del Toro, Elana Smilow, Warren A. Zuckerman, D. George, Kavita Radhakrishnan, Monica Bhatia, Zhezhen Jin, James Garvin, and L.A. Baxter-Lowe
- Subjects
Adult ,Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,Anemia ,medicine.medical_treatment ,Pilot Projects ,Anemia, Sickle Cell ,Hematopoietic stem cell transplantation ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,Young Adult ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Prospective Studies ,Child ,Alemtuzumab ,Busulfan ,Transplantation Chimera ,Transplantation ,business.industry ,Siblings ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,Tissue Donors ,Fludarabine ,Regimen ,Child, Preschool ,Immunology ,Female ,business ,Vidarabine ,medicine.drug - Abstract
BU and CY (BU/CY; 200 mg/kg) before HLA-matched sibling allo-SCT in children with sickle cell disease (SCD) is associated with ~85% EFS but is limited by the acute and late effects of BU/CY myeloablative conditioning. Alternatives include reduced toxicity but more immunosuppressive conditioning. We investigated in a prospective single institutional study, the safety and efficacy of a reduced-toxicity conditioning (RTC) regimen of BU 12.8-16 mg/kg, fludarabine 180 mg/m(2), alemtuzumab 54 mg/m(2) (BFA) before HLA-matched sibling donor transplantation in pediatric recipients with symptomatic SCD. Eighteen patients, median age 8.9 years (2.3-20.2), M/F 15/3, 15 sibling BM and 3 sibling cord blood (CB) were transplanted. Mean whole blood and erythroid donor chimerism was 91% and 88%, at days +100 and +365, respectively. Probability of grade II-IV acute GVHD was 17%. Two-year EFS and OS were both 100%. Neurological, pulmonary and cardiovascular function were stable or improved at 2 years. BFA RTC and HLA-matched sibling BM and CB allo-SCT in pediatric recipients result in excellent EFS, long-term donor chimerism, low incidence of GVHD and stable/improved organ function.
- Published
- 2014
- Full Text
- View/download PDF
50. Inotuzumab Ozogamicin Is an Effective Salvage Therapy in Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia with High-Risk Molecular Features, Including TP53 Loss
- Author
-
Charlene C. Kabel, Christopher J. Forlenza, Xiaochuan Yang, Jae H. Park, Mark B. Geyer, and Amber C. King
- Subjects
Inotuzumab ozogamicin ,Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Ponatinib ,Salvage therapy ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Immunotherapy ,Biochemistry ,Chemotherapy regimen ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,Vindesine ,Blinatumomab ,business ,medicine.drug - Abstract
Introduction: Adults with (w/) B-cell acute lymphoblastic leukemia (B-ALL) exhibit high rates of complete response (CR) to induction chemotherapy, but relapse is common. Inotuzumab ozogamicin (IO), an antibody-drug conjugate targeting CD22, achieves high rates of CR in patients (pts) w/ relapsed/refractory (R/R) B-ALL and is FDA-approved for R/R B-ALL in adults. It remains unknown whether cytogenetic and molecular features associated w/ decreased response rate and poor prognosis following conventional chemotherapy are associated w/ response to IO. As such, we investigated the relationship between several high-risk genetic alterations and outcome following IO treatment in pts w/ R/R B-ALL. Methods: We reviewed electronic medical records of pts of all ages w/ R/R B-ALL or chronic myeloid leukemia in lymphoid blast phase (CML-LBP) receiving IO at Memorial Sloan Kettering Cancer Center (MSK) between January 2011 and April 2019. The primary objective was to assess whether recurrent cytogenetic or molecular features were associated w/ achievement of CR or CR w/ incomplete hematologic recovery (CRi), w/ or w/o measurable residual disease (MRD), and disease-free (DFS) and overall survival (OS) following IO. Secondary objectives included association of baseline clinical features, including central nervous system (CNS) or other extramedullary (EM) disease, w/ outcomes post-IO. MRD was defined as any unequivocal evidence of B-ALL detectable by RT-PCR (Ph+ ALL) or flow cytometry (FACS). Genomic alterations were defined by MSK IMPACT-Heme (Cheng, J Mol Diagn, 2015), FoundationOne Heme, or similar platforms. A set of selected high-risk (HR) features in Philadelphia chromosome-negative (Ph-) B-ALL was defined prior to the analysis (HR: mutations/loss of TP53, IKZF1/3, CDKN2A, CREBBP; activating RAS mutations; "Ph-like" profile). DFS and OS were computed using Kaplan-Meier methods and compared between groups using log-tank tests. Results: 32 pts (13F, 19M) w/ R/R B-ALL (n=31) or CML-LBP (n=1) treated w/ IO were identified. IO was given as monotherapy in 27 pts and w/ other systemic therapy in 5 pts (mini-hyper-CVD-like regimen, n=4; ponatinib, n=1). Median age at start of IO was 45 years (range 3-78). 10 pts had undergone prior allogeneic hematopoietic cell transplantation (alloHCT). Seven and 15 pts had a history of CNS disease or other EM involvement by B-ALL, respectively, including 3 and 6 pts immediately prior to IO, respectively. Pts received a median 3 lines of salvage prior to IO, including prior CD19-targeted immunotherapy (blinatumomab and/or CAR-T cells) in 24 pts(Table 1). Among 27 pts w/ Ph- B-ALL, 12 had the selected HR features (Table 2). Five pts had Ph+ ALL (n=4) or CML-LBP (n=1) and 5/5 harbored ABL1 kinase domain point mutations (4/5 w/ T315I mutation). 22 pts had at least one successful molecular profiling panel.29 patients had initial cytogenetic studies, of whom 28 patients had evaluable karyotypes. 23 pts had best response to IO of CR/CRi (MRD-, n=15; MRD+, n=8). 9 pts had no objective response to ≥1 cycle of IO. Of the 12 Ph- pts w/ selected HR mutations, 11 achieved CR/CRi. Notably, 6/6 pts w/ TP53 mutation/deletion and 5/5 pts w/ IKZF1/3 mutations (3/3 pts w/ both TP53 & IKZF mutations) achieved CR/CRi. Both pts w/ Ras mutations and 2/3 w/ Ph-like B-ALL achieved CR/CRi. 7/11 HR responders underwent alloHCT post-IO (3 had undergone pre-IO alloHCT). Pts w/ Ph- B-ALL w/ HR mutations demonstrated similar CR/CRi rate and OS to pts w/ Ph- B-ALL w/o defined HR mutations (Fig 1A-B). In contrast, only 1/5 pts w/ Ph+ ALL achieved CR/CRi (was MRD+) and 4/5 showed persistent B-ALL. OS was superior among pts w/ Ph- vs Ph+ B-ALL post-IO (8.0 vs 1.9 months, p=0.0068, Fig 1C). Among pts w/ EM disease immediately prior to IO, 3/6 achieved CR/CRi, including CR in 1 pt w/ a cardiac mass. Median DFS was 3.2 months vs. not reached following achievement of MRD+ vs MRD- CR, respectively (p=ns, Fig 1D). Conclusions: HR molecular features associated w/ poor response to chemotherapy were not associated w/ inferior response rate and overall prognosis following IO in this small series. Notably, pts w/ Ph+ ALL (all w/ ABL1 mutations) exhibited suboptimal response, possibly as pts received IO only in advanced disease states following TKI failure. This small report supports investigation of IO in frontline therapy for pts w/ B-ALL w/ HR mutations to spare unnecessary toxicities of chemotherapy and bridge successfully to alloHCT. Disclosures King: Genentech: Other: Advisory Board ; Astrazeneca: Other: Advisory board; Incyte: Other: Advisory Board. Park:Allogene: Consultancy; Amgen: Consultancy; AstraZeneca: Consultancy; Autolus: Consultancy; GSK: Consultancy; Incyte: Consultancy; Kite Pharma: Consultancy; Novartis: Consultancy; Takeda: Consultancy. Geyer:Dava Oncology: Honoraria; Amgen: Research Funding. OffLabel Disclosure: Inotuzumab ozogamicin is not FDA approved for pediatric patients with relapsed/refractory B-cell acute lymphoblastic leukemia.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.