71 results on '"Geoffrey Fell"'
Search Results
2. Clonal Hematopoiesis in Young Women Treated for Breast Cancer
- Author
-
Christopher J. Gibson, Geoffrey Fell, Tal Sella, Adam S. Sperling, Craig Snow, Shoshana M. Rosenberg, Greg Kirkner, Ashka Patel, Deborah Dillon, Alexander G. Bick, Donna Neuberg, Ann H. Partridge, and Peter G. Miller
- Subjects
Cancer Research ,Oncology - Abstract
Background: Young women treated for breast cancer with cytotoxic therapies are at risk for clonal hematopoiesis of indeterminate potential (CHIP), a condition in which blood cells carrying a somatic mutation associated with hematologic malignancy comprise at least 4% of the total blood system. CHIP has primarily been studied in older patient cohorts with limited clinical phenotyping. Materials and Methods: We performed targeted sequencing on longitudinal blood samples to characterize the clonal hematopoietic landscape of 878 women treated for breast cancer enrolled in the prospective Young Women's Breast Cancer Study. Results: We identified somatic driver mutations in 252 study subjects (28.7%), but only 24 (2.7%) had clones large enough to meet criteria for CHIP. The most commonly mutated genes were DNMT3A and TET2, similar to mutations observed in noncancer cohorts. At 9-year median follow-up, we found no association between the presence of a somatic blood mutation (regardless of clone size) and adverse breast cancer (distant relapse-free survival) or non–breast cancer-related outcomes in this cohort. A subset of paired blood samples obtained over 4 years showed no evidence of mutant clonal expansion, regardless of genotype. Finally, we identified a subset of patients with likely germline mutations in genes known to contribute to inherited cancer risk, such as TP53 and ATM. Conclusions: Our data show that for young women with early-stage breast cancer, CHIP is uncommon after cytotoxic exposure, is unlikely to contribute to adverse outcomes over the decade-long follow-up and may not require additional monitoring if discovered incidentally.
- Published
- 2023
3. In VivoModeling of CLL Transformation to Richter Syndrome Reveals Convergent Evolutionary Paths and Therapeutic Vulnerabilities
- Author
-
Elisa ten Hacken, Tomasz Sewastianik, Shanye Yin, Gabriela Brunsting Hoffmann, Michaela Gruber, Kendell Clement, Livius Penter, Robert A. Redd, Neil Ruthen, Sébastien Hergalant, Alanna Sholokhova, Geoffrey Fell, Erin M. Parry, Julien Broséus, Romain Guieze, Fabienne Lucas, María Hernández-Sánchez, Kaitlyn Baranowski, Jackson Southard, Heather Joyal, Leah Billington, Fara Faye D. Regis, Elizabeth Witten, Mohamed Uduman, Binyamin A. Knisbacher, Shuqiang Li, Haoxiang Lyu, Tiziana Vaisitti, Silvia Deaglio, Giorgio Inghirami, Pierre Feugier, Stephan Stilgenbauer, Eugen Tausch, Matthew S. Davids, Gad Getz, Kenneth J. Livak, Ivana Bozic, Donna S. Neuberg, Ruben D. Carrasco, and Catherine J. Wu
- Subjects
General Medicine - Abstract
Transformation to aggressive disease histologies generates formidable clinical challenges across cancers, but biological insights remain few. We modeled the genetic heterogeneity of chronic lymphocytic leukemia (CLL) through multiplexed in vivo CRISPR-Cas9 B-cell editing of recurrent CLL loss-of-function drivers in mice and recapitulated the process of transformation from indolent CLL into large cell lymphoma [i.e., Richter syndrome (RS)]. Evolutionary trajectories of 64 mice carrying diverse combinatorial gene assortments revealed coselection of mutations in Trp53, Mga, and Chd2 and the dual impact of clonal Mga/Chd2 mutations on E2F/MYC and interferon signaling dysregulation. Comparative human and murine RS analyses demonstrated tonic PI3K signaling as a key feature of transformed disease, with constitutive activation of the AKT and S6 kinases, downmodulation of the PTEN phosphatase, and convergent activation of MYC/PI3K transcriptional programs underlying enhanced sensitivity to MYC/mTOR/PI3K inhibition. This robust experimental system presents a unique framework to study lymphoid biology and therapy.Significance:Mouse models reflective of the genetic complexity and heterogeneity of human tumors remain few, including those able to recapitulate transformation to aggressive disease histologies. Herein, we model CLL transformation into RS through multiplexed in vivo gene editing, providing key insight into the pathophysiology and therapeutic vulnerabilities of transformed disease.This article is highlighted in the In This Issue feature, p. 101
- Published
- 2022
4. Peripheral blood <scp>CD3</scp> + T‐cell gene expression biomarkers correlate with clinical frailty in patients with haematological malignancies
- Author
-
Geoffrey Fell, Ashley E. Rosko, Gregory A. Abel, Clark Dumontier, Kelly J. Higby, Anays Murillo, Donna S. Neuberg, Christin E. Burd, and Andrew A. Lane
- Subjects
Hematology - Published
- 2022
5. Supplemental Fig. S3 from Clonal Hematopoiesis in Young Women Treated for Breast Cancer
- Author
-
Peter G. Miller, Ann H. Partridge, Donna Neuberg, Alexander G. Bick, Deborah Dillon, Ashka Patel, Greg Kirkner, Shoshana M. Rosenberg, Craig Snow, Adam S. Sperling, Tal Sella, Geoffrey Fell, and Christopher J. Gibson
- Abstract
Longitudinal Mutation Analysis in the YWS
- Published
- 2023
6. Data from Clonal Hematopoiesis in Young Women Treated for Breast Cancer
- Author
-
Peter G. Miller, Ann H. Partridge, Donna Neuberg, Alexander G. Bick, Deborah Dillon, Ashka Patel, Greg Kirkner, Shoshana M. Rosenberg, Craig Snow, Adam S. Sperling, Tal Sella, Geoffrey Fell, and Christopher J. Gibson
- Abstract
Background:Young women treated for breast cancer with cytotoxic therapies are at risk for clonal hematopoiesis of indeterminate potential (CHIP), a condition in which blood cells carrying a somatic mutation associated with hematologic malignancy comprise at least 4% of the total blood system. CHIP has primarily been studied in older patient cohorts with limited clinical phenotyping.Materials and Methods:We performed targeted sequencing on longitudinal blood samples to characterize the clonal hematopoietic landscape of 878 women treated for breast cancer enrolled in the prospective Young Women's Breast Cancer Study.Results:We identified somatic driver mutations in 252 study subjects (28.7%), but only 24 (2.7%) had clones large enough to meet criteria for CHIP. The most commonly mutated genes were DNMT3A and TET2, similar to mutations observed in noncancer cohorts. At 9-year median follow-up, we found no association between the presence of a somatic blood mutation (regardless of clone size) and adverse breast cancer (distant relapse-free survival) or non–breast cancer-related outcomes in this cohort. A subset of paired blood samples obtained over 4 years showed no evidence of mutant clonal expansion, regardless of genotype. Finally, we identified a subset of patients with likely germline mutations in genes known to contribute to inherited cancer risk, such as TP53 and ATM.Conclusions:Our data show that for young women with early-stage breast cancer, CHIP is uncommon after cytotoxic exposure, is unlikely to contribute to adverse outcomes over the decade-long follow-up and may not require additional monitoring if discovered incidentally.
- Published
- 2023
7. Supplemental Table S5 from Clonal Hematopoiesis in Young Women Treated for Breast Cancer
- Author
-
Peter G. Miller, Ann H. Partridge, Donna Neuberg, Alexander G. Bick, Deborah Dillon, Ashka Patel, Greg Kirkner, Shoshana M. Rosenberg, Craig Snow, Adam S. Sperling, Tal Sella, Geoffrey Fell, and Christopher J. Gibson
- Abstract
Cohort Characteristics Broken Down by CH (Any VAF)
- Published
- 2023
8. Data from In Vivo Modeling of CLL Transformation to Richter Syndrome Reveals Convergent Evolutionary Paths and Therapeutic Vulnerabilities
- Author
-
Catherine J. Wu, Ruben D. Carrasco, Donna S. Neuberg, Ivana Bozic, Kenneth J. Livak, Gad Getz, Matthew S. Davids, Eugen Tausch, Stephan Stilgenbauer, Pierre Feugier, Giorgio Inghirami, Silvia Deaglio, Tiziana Vaisitti, Haoxiang Lyu, Shuqiang Li, Binyamin A. Knisbacher, Mohamed Uduman, Elizabeth Witten, Fara Faye D. Regis, Leah Billington, Heather Joyal, Jackson Southard, Kaitlyn Baranowski, María Hernández-Sánchez, Fabienne Lucas, Romain Guieze, Julien Broséus, Erin M. Parry, Geoffrey Fell, Alanna Sholokhova, Sébastien Hergalant, Neil Ruthen, Robert A. Redd, Livius Penter, Kendell Clement, Michaela Gruber, Gabriela Brunsting Hoffmann, Shanye Yin, Tomasz Sewastianik, and Elisa ten Hacken
- Abstract
Transformation to aggressive disease histologies generates formidable clinical challenges across cancers, but biological insights remain few. We modeled the genetic heterogeneity of chronic lymphocytic leukemia (CLL) through multiplexed in vivo CRISPR-Cas9 B-cell editing of recurrent CLL loss-of-function drivers in mice and recapitulated the process of transformation from indolent CLL into large cell lymphoma [i.e., Richter syndrome (RS)]. Evolutionary trajectories of 64 mice carrying diverse combinatorial gene assortments revealed coselection of mutations in Trp53, Mga, and Chd2 and the dual impact of clonal Mga/Chd2 mutations on E2F/MYC and interferon signaling dysregulation. Comparative human and murine RS analyses demonstrated tonic PI3K signaling as a key feature of transformed disease, with constitutive activation of the AKT and S6 kinases, downmodulation of the PTEN phosphatase, and convergent activation of MYC/PI3K transcriptional programs underlying enhanced sensitivity to MYC/mTOR/PI3K inhibition. This robust experimental system presents a unique framework to study lymphoid biology and therapy.Significance:Mouse models reflective of the genetic complexity and heterogeneity of human tumors remain few, including those able to recapitulate transformation to aggressive disease histologies. Herein, we model CLL transformation into RS through multiplexed in vivo gene editing, providing key insight into the pathophysiology and therapeutic vulnerabilities of transformed disease.This article is highlighted in the In This Issue feature, p. 101
- Published
- 2023
9. Supplementary Figures from In Vivo Modeling of CLL Transformation to Richter Syndrome Reveals Convergent Evolutionary Paths and Therapeutic Vulnerabilities
- Author
-
Catherine J. Wu, Ruben D. Carrasco, Donna S. Neuberg, Ivana Bozic, Kenneth J. Livak, Gad Getz, Matthew S. Davids, Eugen Tausch, Stephan Stilgenbauer, Pierre Feugier, Giorgio Inghirami, Silvia Deaglio, Tiziana Vaisitti, Haoxiang Lyu, Shuqiang Li, Binyamin A. Knisbacher, Mohamed Uduman, Elizabeth Witten, Fara Faye D. Regis, Leah Billington, Heather Joyal, Jackson Southard, Kaitlyn Baranowski, María Hernández-Sánchez, Fabienne Lucas, Romain Guieze, Julien Broséus, Erin M. Parry, Geoffrey Fell, Alanna Sholokhova, Sébastien Hergalant, Neil Ruthen, Robert A. Redd, Livius Penter, Kendell Clement, Michaela Gruber, Gabriela Brunsting Hoffmann, Shanye Yin, Tomasz Sewastianik, and Elisa ten Hacken
- Abstract
Supplementary file contains a PDF version of supplementary figures S1-S7 and associated figure legends.
- Published
- 2023
10. Supplementary Tables from In Vivo Modeling of CLL Transformation to Richter Syndrome Reveals Convergent Evolutionary Paths and Therapeutic Vulnerabilities
- Author
-
Catherine J. Wu, Ruben D. Carrasco, Donna S. Neuberg, Ivana Bozic, Kenneth J. Livak, Gad Getz, Matthew S. Davids, Eugen Tausch, Stephan Stilgenbauer, Pierre Feugier, Giorgio Inghirami, Silvia Deaglio, Tiziana Vaisitti, Haoxiang Lyu, Shuqiang Li, Binyamin A. Knisbacher, Mohamed Uduman, Elizabeth Witten, Fara Faye D. Regis, Leah Billington, Heather Joyal, Jackson Southard, Kaitlyn Baranowski, María Hernández-Sánchez, Fabienne Lucas, Romain Guieze, Julien Broséus, Erin M. Parry, Geoffrey Fell, Alanna Sholokhova, Sébastien Hergalant, Neil Ruthen, Robert A. Redd, Livius Penter, Kendell Clement, Michaela Gruber, Gabriela Brunsting Hoffmann, Shanye Yin, Tomasz Sewastianik, and Elisa ten Hacken
- Abstract
Supplementary file contains all supplementary tables S1-S16 in Excel format in the order in which they appear in the text.
- Published
- 2023
11. Supplementary Data from Deviation from the Proportional Hazards Assumption in Randomized Phase 3 Clinical Trials in Oncology: Prevalence, Associated Factors, and Implications
- Author
-
Brian M. Alexander, Lorenzo Trippa, Alyssa M. Vanderbeek, Andrea Arfé, Steffen Ventz, Geoffrey Fell, and Rifaquat Rahman
- Abstract
Supplementary text, figures and tables
- Published
- 2023
12. Supplementary Data from Pathologic Complete Response after Neoadjuvant Chemotherapy and Impact on Breast Cancer Recurrence and Survival: A Comprehensive Meta-analysis
- Author
-
Aditya Bardia, Lorenzo Trippa, Giovanni Parmigiani, Steven J. Isakoff, Beverly Moy, Brian Alexander, Barbara L. Smith, Kerry L. Reynolds, Rachel Greenup, Chandni Sharma, Andrea Arfe, Geoffrey Fell, and Laura M. Spring
- Abstract
Online supplement
- Published
- 2023
13. Data from Pathologic Complete Response after Neoadjuvant Chemotherapy and Impact on Breast Cancer Recurrence and Survival: A Comprehensive Meta-analysis
- Author
-
Aditya Bardia, Lorenzo Trippa, Giovanni Parmigiani, Steven J. Isakoff, Beverly Moy, Brian Alexander, Barbara L. Smith, Kerry L. Reynolds, Rachel Greenup, Chandni Sharma, Andrea Arfe, Geoffrey Fell, and Laura M. Spring
- Abstract
Purpose:While various studies have highlighted the prognostic significance of pathologic complete response (pCR) after neoadjuvant chemotherapy (NAT), the impact of additional adjuvant therapy after pCR is not known.Experimental Design:PubMed was searched for studies with NAT for breast cancer and individual patient-level data was extracted for analysis using plot digitizer software. HRs, with 95% probability intervals (PI), measuring the association between pCR and overall survival (OS) or event-free survival (EFS), were estimated using Bayesian piece-wise exponential proportional hazards hierarchical models including pCR as predictor.Results:Overall, 52 of 3,209 publications met inclusion criteria, totaling 27,895 patients. Patients with a pCR after NAT had significantly better EFS (HR = 0.31; 95% PI, 0.24–0.39), particularly for triple-negative (HR = 0.18; 95% PI, 0.10–0.31) and HER2+ (HR = 0.32; 95% PI, 0.21–0.47) disease. Similarly, pCR after NAT was also associated with improved survival (HR = 0.22; 95% PI, 0.15–0.30). The association of pCR with improved EFS was similar among patients who received subsequent adjuvant chemotherapy (HR = 0.36; 95% PI, 0.19–0.67) and those without adjuvant chemotherapy (HR = 0.36; 95% PI, 0.27–0.54), with no significant difference between the two groups (P = 0.60).Conclusions:Achieving pCR following NAT is associated with significantly better EFS and OS, particularly for triple-negative and HER2+ breast cancer. The similar outcomes with or without adjuvant chemotherapy in patients who attain pCR likely reflects tumor biology and systemic clearance of micrometastatic disease, highlighting the potential of escalation/deescalation strategies in the adjuvant setting based on neoadjuvant response.See related commentary by Esserman, p. 2771
- Published
- 2023
14. Data from Deviation from the Proportional Hazards Assumption in Randomized Phase 3 Clinical Trials in Oncology: Prevalence, Associated Factors, and Implications
- Author
-
Brian M. Alexander, Lorenzo Trippa, Alyssa M. Vanderbeek, Andrea Arfé, Steffen Ventz, Geoffrey Fell, and Rifaquat Rahman
- Abstract
Purpose:Deviations from proportional hazards (DPHs), which may be more prevalent in the era of precision medicine and immunotherapy, can lead to underpowered trials or misleading conclusions. We used a meta-analytic approach to estimate DPHs across cancer trials, investigate associated factors, and evaluate data-analysis approaches for future trials.Experimental Design: We searched PubMed for phase III trials in breast, lung, prostate, and colorectal cancer published in a preselected list of journals between 2014 and 2016 and extracted individual patient-level data (IPLD) from Kaplan–Meier curves. We re-analyzed IPLD to identify DPHs. Potential efficiency gains, when DPHs were present, of alternative statistical methods relative to standard log-rank based analysis were expressed as sample-size requirements for a fixed power level.Results:From 152 trials, we obtained IPLD on 129,401 patients. Among 304 Kaplan–Meier figures, 75 (24.7%) exhibited evidence of DPHs, including eight of 14 (57%) KM pairs from immunotherapy trials. Trial type [immunotherapy, odds ratio (OR), 4.29; 95% confidence interval (CI), 1.11–16.6], metastatic patient population (OR, 3.18; 95% CI, 1.26–8.05), and non-OS endpoints (OR, 3.23; 95% CI, 1.79–5.88) were associated with DPHs. In immunotherapy trials, alternative statistical approaches allowed for more efficient clinical trials with fewer patients (up to 74% reduction) relative to log-rank testing.Conclusions:DPHs were found in a notable proportion of time-to-event outcomes in published clinical trials in oncology and was more common for immunotherapy trials and non-OS endpoints. Alternative statistical methods, without proportional hazards assumptions, should be considered in the design and analysis of clinical trials when the likelihood of DPHs is high.
- Published
- 2023
15. Maintenance Therapy with Venetoclax/Azacitidine Can be Safely Given after Venetoclax/FluBu2 RIC Allogeneic Transplantation for the Treatment of High Risk MDS/AML: Results of a Phase 1 Study
- Author
-
Jacqueline S. Garcia, Haesook T Kim, Jennifer Brock, H. Moses Murdock, Corey S. Cutler, Daniel J. DeAngelo, Christopher J. Gibson, Mahasweta Gooptu, Vincent Ho, John Koreth, Marlise R. Luskin, Sarah Nikiforow, Rizwan Romee, Roman M Shapiro, Richard M. Stone, Martha Wadleigh, Eric S. Winer, Michela Ansuinelli, Eliza Elliot, Geoffrey Fell, Hannah Karp, Jeremy Ryan, Anthony G. Letai, Coleman Lindsley, Robert J Soiffer, Joseph H. Antin, Fiona Loschi, and Jerome Ritz
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
16. Clonal hematopoiesis in patients receiving chimeric antigen receptor T-cell therapy
- Author
-
Nikhil C. Munshi, Christopher J. Gibson, Caron A. Jacobson, Peter Miller, Max Jan, Mark B. Leick, Geoffrey Fell, Benjamin L. Ebert, Marcela V. Maus, Elliott J. Brea, Yu-Tzu Tai, Satyen H. Gohil, Donna Neuberg, Catherine J. Wu, and Adam S. Sperling
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Cell- and Tissue-Based Therapy ,Immunotherapy, Adoptive ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Internal medicine ,medicine ,Humans ,Autologous transplantation ,Cytotoxic T cell ,Multiple myeloma ,Receptors, Chimeric Antigen ,business.industry ,Lymphoma, Non-Hodgkin ,Hematology ,Middle Aged ,medicine.disease ,Stimulus Report ,Chimeric antigen receptor ,Lymphoma ,Cytokine release syndrome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Chimeric Antigen Receptor T-Cell Therapy ,Clonal Hematopoiesis ,business - Abstract
Chimeric antigen receptor (CAR) T-cells have emerged as an efficacious modality in patients with non-Hodgkin lymphoma (NHL) and multiple myeloma (MM). Clonal hematopoiesis of indeterminate potential (CHIP), a state in which mutations in hematopoietic cells give rise to a clonal population of cells, is more common in patients exposed to cytotoxic therapies, has been shown to influence inflammatory immune programs, and is associated with an adverse prognosis in patients with NHL and MM receiving autologous transplantation. We therefore hypothesized that CHIP could influence clinical outcomes in patients receiving CAR T-cell therapy. In a cohort of 154 patients with NHL or MM receiving CAR T-cells, we found that CHIP was present in 48% of patients and associated with increased rates of complete response and cytokine release syndrome severity, but only in patients younger than age 60 years. Despite these differences, CHIP was not associated with a difference in progression-free or overall survival, regardless of age. Our data suggest that CHIP can influence CAR T-cell biology and clinical outcomes, but, in contrast to autologous transplantation, CHIP was not associated with worse survival and should not be a reason to exclude individuals from receiving this potentially life-prolonging treatment.
- Published
- 2021
17. Peripheral blood CD3
- Author
-
Geoffrey, Fell, Ashley E, Rosko, Gregory A, Abel, Clark, Dumontier, Kelly J, Higby, Anays, Murillo, Donna S, Neuberg, Christin E, Burd, and Andrew A, Lane
- Subjects
Adult ,Aged, 80 and over ,CD28 Antigens ,Frailty ,Receptors, Aryl Hydrocarbon ,Frail Elderly ,Hematologic Neoplasms ,T-Lymphocytes ,Gene Expression ,Humans ,Middle Aged ,Biomarkers ,Aged - Abstract
Older patients with cancer often receive treatment regimens based on their age without considering other objective factors that may influence outcomes. Assessment of frailty can identify older patients who are robust and therefore more likely to benefit from intensive treatment, or conversely, frail and might instead be offered alternative approaches. However, such assessment requires specialised training and dedicated clinical resources. Alternative quantitative biomarkers associated with frailty are lacking. Here, we asked if expression signatures of 74 immune cell, ageing, and senescence-related messenger RNAs in purified peripheral blood T cells could identify associations with clinical frailty in patients with haematological malignancies. We studied 69 patients between the ages of 36 and 92 years (median 76 years) with leukaemia, lymphoma, or multiple myeloma, across two institutions. Expression of four genes (aryl hydrocarbon receptor [AHR], CD27, CD28, and interleukin-2 receptor subunit alpha [IL2RA; CD25]) in T cells was associated with frailty, independent of age. An expression-based regression model had 76% sensitivity and 90% specificity to assign a patient as robust. These data identify measurable peripheral blood correlates of clinical frailty and suggest biomarkers for future prospective assessment.
- Published
- 2022
18. Intracranial, intratumoral implantation of drug-releasing microdevices in patients with high grade gliomas is feasible, safe, and may predict tumor response to systemic chemotherapy
- Author
-
Pierpaolo Peruzzi, Christine Dominas, Geoffrey Fell, Sarah Blitz, Joshua Bernstock, Hassan Dawood, Daniel Triggs, Sebastian Ahn, Sharat Bhagavatula, Zuzana Tatarova, Michael Pannel, Kyla Truman, Anna Ball, E. Chiocca, Keith Ligon, Patrick Wen, and Oliver Jonas
- Abstract
The lack of reliable predictive biomarkers to guide effective therapy is a major obstacle for the advancement of therapy for high grade gliomas (HGG), and particularly glioblastoma (GBM), one of the few cancers whose prognosis has not improved over the past several decades. With this pilot clinical trial we provide first in human evidence that drug-releasing intratumoral microdevices (IMD) can be safely and effectively used to obtain patient-specific, high throughput molecular and histopathological data to inform selection of drugs based on their observed antitumor effect in situ. The use of IMD is seamlessly integrated in standard surgical practice during tumor resection. None of the six enrolled patients experienced adverse events related to the IMD, and the retrieved tissue was usable for downstream analysis for 11 out of 12 retrieved specimens. Molecular analysis of the specimens provided, for the first time in humans, preliminary evidence of the robustness of the readout, with strong correlation between IMD analysis and clinic-radiological responses to temozolomide. From an investigational aspect, the amount of information obtained with IMD allows unprecedented characterization of tissue effects of any drugs of interest, within the physiological context of the intact tumor.
- Published
- 2022
19. A quantitative framework for modeling COVID-19 risk during adjuvant therapy using published randomized trials of glioblastoma in the elderly
- Author
-
Shyam K. Tanguturi, Rifaquat Rahman, Brian M. Alexander, Steffen Ventz, Daniel N. Cagney, Patrick Y. Wen, Shervin Tabrizi, Geoffrey Fell, and Lorenzo Trippa
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Clinical Neurology ,elderly ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,Pandemic ,medicine ,Adjuvant therapy ,AcademicSubjects/MED00300 ,Temozolomide ,business.industry ,Risk of infection ,Hazard ratio ,COVID-19 ,Cancer ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,randomized controlled trials ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Glioblastoma ,business ,Fast-Track Article ,Adjuvant ,medicine.drug - Abstract
Background During the ongoing COVID-19 pandemic, contact with the health care system for cancer treatment can increase risk of infection and associated mortality. Treatment recommendations must consider this risk for elderly and vulnerable cancer patients. We reanalyzed trials in elderly glioblastoma (GBM) patients, incorporating COVID-19 risk, in order to provide a quantitative framework for comparing different radiation (RT) fractionation schedules on patient outcomes. Methods We extracted individual patient-level data for 1321 patients from Kaplan–Meier curves from 5 randomized trials on treatment of elderly GBM patients including available subanalyses based on O6-methylguanine-DNA methyltransferase (MGMT) methylation status. We simulated trial data with incorporation of COVID-19–associated mortality risk in several scenarios (low, medium, and high infection and mortality risks). Median overall survival and hazard ratios were calculated for each simulation replicate. Results Our simulations reveal how COVID-19–associated risks affect survival under different treatment regimens. Hypofractionated RT with concurrent and adjuvant temozolomide (TMZ) demonstrated the best outcomes in low and medium risk scenarios. In frail elderly patients, shorter courses of RT are preferable. In patients with methylated MGMT receiving single modality treatment, TMZ-alone treatment approaches may be an option in settings with high COVID-19–associated risk. Conclusions Incorporation of COVID-19–associated risk models into analysis of randomized trials can help guide clinical decisions during this pandemic. In elderly GBM patients, our results support prioritization of hypofractionated RT and highlight the utility of MGMT methylation status in decision making in pandemic scenarios. Our quantitative framework can serve as a model for assessing COVID-19 risk associated with treatment across neuro-oncology. Key Points • Re-analysis of randomized controlled trials in COVID-19 era gives insight on optimal treatment of GBM. • Hypofractionated RT or temozolomide alone may be reasonable options in high risk pandemic settings. • A quantitative framework incorporating COVID-19 risks can be applied across neuro-oncology.
- Published
- 2020
20. Comparison of demographics, disease characteristics, and outcomes between Black and White patients with myelodysplastic syndromes: A population-based study
- Author
-
Arnaud, Lesegretain, Andrew, Brunner, Andrew J, King, Abderrahmane, Laadem, Geoffrey, Fell, and Amir T, Fathi
- Subjects
Cancer Research ,Oncology ,Hematology - Abstract
Racial disparities in cancer care and outcomes have been well documented in various malignancies, with Black patients having the highest death rate and shortest survival of any racial/ethnic group in the United States (US) for most cancers. However, there have been limited studies on racial/ethnic disparities in myelodysplastic syndromes (MDS). Our study characterized and compared differences in baseline demographics, clinical characteristics, socioeconomic factors, and overall survival (OS) between Black and White patients with MDS in the US. We used the Surveillance, Epidemiology, and End Results (SEER) Program and included 37,562 patients (Black, 8.1 %; White, 91.9 %) diagnosed between 2001 and 2013. We observed significant differences in baseline characteristics between cohorts. In a univariate analysis, Black race was associated with longer survival (hazard ratio [HR]: 0.83; 95 % confidence interval [CI], 0.79-0.86; p 0.001). The association between race and survival was attenuated but remained significant in various models to adjust for differences in baseline characteristics (HR in multivariable analysis, 0.92; 95 % CI, 0.87-0.96); p 0.001). Subgroup analysis by histology revealed differences in the association between race and OS. Refractory anemia (RA), RA with ring sideroblasts, and MDS-not otherwise specified, a category in SEER representing a poorly defined MDS subset for 52 % of cases in our study, favored Black patients. RA with excess blasts favored White patients. The overall finding that Black race is associated with better OS outcomes, when compared with White patients, needs to be interpreted with caution and nuanced by histology. Additional research to explore these associations is warranted.
- Published
- 2023
21. Increased mitochondrial apoptotic priming with targeted therapy predicts clinical response to re‐induction chemotherapy
- Author
-
Donna Neuberg, Michael Burgess, Karen K. Ballen, Steven A. Carr, Geoffrey Fell, Benjamin L. Ebert, Anthony Letai, Jacqueline S. Garcia, Binyam Yilma, Richard Stone, Andrew M. Brunner, Hasmik Keshishian, Adam S. Sperling, Daniel J. DeAngelo, Bruno C. Medeiros, and Shruti Bhatt
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Myeloid ,medicine.medical_treatment ,Apoptosis ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Lenalidomide ,Etoposide ,Aged ,Chemotherapy ,business.industry ,Cytarabine ,Induction chemotherapy ,Induction Chemotherapy ,Hematology ,Middle Aged ,medicine.disease ,Mitochondria ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Mitoxantrone ,business ,030215 immunology ,medicine.drug - Abstract
Most patients with relapsed or refractory (R/R) acute myeloid leukemia (AML) do not benefit from current re-induction or approved targeted therapies. In the absence of targetable genetic mutations, there is minimal guidance on optimal treatment selection particularly in the R/R setting highlighting an unmet need for clinically useful functional biomarkers. Blood and bone marrow samples from patients treated on two clinical trials were used to test the combination of lenalidomide (LEN) and MEC (mitoxantrone, etoposide, and cytarabine) chemotherapy in R/R AML patients. The bone marrow samples were available to test the clinical utility of the mitochondrial apoptotic BH3 and dynamic BH3 profiling (DBP) assays in predicting response, as there was no clear genetic biomarker identifying responders. To test whether LEN-induced mitochondrial priming predicted clinical response to LEN-MEC therapy, we performed DBP on patient myeloblasts. We found that short-term ex vivo treatment with lenalidomide discriminated clinical responders from non-responders based on drug-induced change in priming (delta priming). Using paired patient samples collected before and after clinical LEN treatment (prior to MEC dosing), we confirmed LEN-induced increased apoptotic priming in vivo, suggesting LEN enhanced vulnerability of myeloblasts to cytotoxic MEC chemotherapy. This is the first study demonstrating the potential role of DBP in predicting clinical response to a combination regimen. Our findings demonstrate that functional properties of relapsed AML can identify active therapies.
- Published
- 2019
22. Historical expectations with DNA methyltransferase inhibitor monotherapy in MDS: when is combination therapy truly 'promising'?
- Author
-
Andrew M. Brunner, Geoffrey Fell, and David P. Steensma
- Subjects
Antimetabolites, Antineoplastic ,Motivation ,Treatment Outcome ,Myelodysplastic Syndromes ,Azacitidine ,Humans ,Hematology ,DNA ,Methyltransferases ,Enzyme Inhibitors ,Decitabine - Abstract
DNA methyltransferase inhibitors (DNMTIs) for patients with higher risk myelodysplastic syndromes (HR-MDS) have low complete remission rates and are not curative. Early DNMTI combination clinical trials in HR-MDS are often termed “promising,” but many randomized trials subsequently failed to show benefit. Clearer understanding of when a combination is likely to improve upon DNMTI monotherapy would inform randomized studies. We reviewed MDS azacitidine or decitabine monotherapy studies. We collected baseline demographics including International Prognostic Scoring System (IPSS) risk, DNMTI, disease characteristics; and response variables including survival and marrow and hematologic responses. Aggregate estimates across studies were calculated using meta-analyses techniques. Using a binomial design, we estimated the necessary operating characteristics to design a phase 2 study showing improved efficacy of a combination over monotherapy. Among 1908 patients, the overall response rate (ORR) was 24% (n = 464; 95% confidence interval [CI], 0.22-0.26): 267 complete response (CR, 14%), 68 partial response (4%), and 129 marrow complete remission (7%). Among 1604 patients for whom a hematologic response was reported, 476 (30%; 95% CI, 0.27-0.32) reported hematologic improvement (HI). More patients treated with azacitidine achieved HI (38%; 95% CI, 0.35-0.41) compared with decitabine (15%; 95% CI, 0.13-0.19), whereas the marrow ORR rate was higher with decitabine (29%; 95% CI, 0.26-0.33) compared with azacitidine (21%; 95% CI, 0.19-0.23). CR rates were similar between DNMTIs: 13% with azacitidine and 16% with decitabine. Variables that influence MDS response include the specific DNMTI backbone and the distribution of IPSS risk of patients enrolled on a trial. Considering these factors can help identify which early combination approaches are worth assessing in larger randomized trials.
- Published
- 2021
23. Poster: MDS-556 Comparison of Demographics, Disease Characteristics and Outcomes Between Black and White Patients With Myelodysplastic Syndrome: A Population-Based Study
- Author
-
Arnaud Lesegretain, Abderrahmane Laadem, Geoffrey Fell, and Amir T. Fathi
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
24. MDS-556 Comparison of Demographics, Disease Characteristics and Outcomes Between Black and White Patients With Myelodysplastic Syndrome: A Population-Based Study
- Author
-
Arnaud Lesegretain, Abderrahmane Laadem, Geoffrey Fell, and Amir T. Fathi
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
25. Adding venetoclax to fludarabine/busulfan RIC transplant for high-risk MDS and AML is feasible, safe, and active
- Author
-
Robert J. Soiffer, Annette S. Kim, R. Coleman Lindsley, Haesook T. Kim, Anthony Letai, Mahasweta Gooptu, Corey Cutler, Thelma Mashaka, Vincent T. Ho, Jeremy Ryan, Jacqueline S. Garcia, Jennifer Brock, H. Moses Murdock, Richard Stone, Sarah Nikiforow, Hannah Q Karp, John Koreth, Fiona Loschi, Geoffrey Fell, Joseph H. Antin, Fabienne Lucas, Danielle S. Potter, Rizwan Romee, Roman M Shapiro, and Daniel J. DeAngelo
- Subjects
Adult ,medicine.medical_specialty ,Clinical Trials and Observations ,medicine.medical_treatment ,Gastroenterology ,chemistry.chemical_compound ,Maintenance therapy ,Internal medicine ,medicine ,Mucositis ,Humans ,Transplantation, Homologous ,Progression-free survival ,Busulfan ,Chemotherapy ,Sulfonamides ,business.industry ,Venetoclax ,Myelodysplastic syndromes ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Fludarabine ,Leukemia, Myeloid, Acute ,surgical procedures, operative ,chemistry ,business ,Vidarabine ,medicine.drug - Abstract
Key Points Adding venetoclax to FluBu2 reduced-intensity conditioning transplant did not impair engraftment or induce excessive graft-versus-host disease.Monitoring measurable residual disease by ultra-sensitive duplex sequencing revealed complex clonal dynamics before and after transplant., Visual Abstract, Adding the selective BCL-2 inhibitor venetoclax to reduced-intensity conditioning chemotherapy (fludarabine and busulfan [FluBu2]) may enhance antileukemic cytotoxicity and thereby reduce the risk of posttransplant relapse. This phase 1 study investigated the recommended phase 2 dose (RP2D) of venetoclax, a BCL-2 selective inhibitor, when added to FluBu2 in adult patients with high-risk acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and MDS/myeloproliferative neoplasms (MPN) undergoing transplant. Patients received dose-escalated venetoclax (200-400 mg daily starting day −8 for 6-7 doses) in combination with fludarabine 30 mg/m2 per day for 4 doses and busulfan 0.8 mg/kg twice daily for 8 doses on day −5 to day −2 (FluBu2). Transplant related–toxicity was evaluated from the first venetoclax dose on day −8 to day 28. Twenty-two patients were treated. At study entry, 5 patients with MDS and MDS/MPN had 5% to 10% marrow blasts, and 18 (82%) of 22 had a persistent detectable mutation. Grade 3 adverse events included mucositis, diarrhea, and liver transaminitis (n = 3 each). Neutrophil/platelet recovery and acute/chronic graft-versus-host-disease rates were similar to those of standard FluBu2. No dose-limiting toxicities were observed. The RP2D of venetoclax was 400 mg daily for 7 doses. With a median follow-up of 14.7 months (range, 8.6-24.8 months), median overall survival was not reached, and progression-free survival was 12.2 months (95% confidence interval, 6.0-not estimable). In patients with high-risk AML, MDS, and MDS/MPN, adding venetoclax to FluBu2 was feasible and safe. To further address relapse risk, assessment of maintenance therapy after venetoclax plus FluBu2 transplant is ongoing. This study was registered at clinicaltrials.gov as #NCT03613532.
- Published
- 2021
26. Cancer patient survival can be parametrized to improve trial precision and reveal time-dependent therapeutic effects
- Author
-
Deborah, Plana, Geoffrey, Fell, Brian M, Alexander, Adam C, Palmer, and Peter K, Sorger
- Subjects
Clinical Trials as Topic ,Models, Statistical ,Cancer Survivors ,Research Design ,Neoplasms ,Humans ,Kaplan-Meier Estimate ,Proportional Hazards Models - Abstract
Individual participant data (IPD) from oncology clinical trials is invaluable for identifying factors that influence trial success and failure, improving trial design and interpretation, and comparing pre-clinical studies to clinical outcomes. However, the IPD used to generate published survival curves are not generally publicly available. We impute survival IPD from ~500 arms of Phase 3 oncology trials (representing ~220,000 events) and find that they are well fit by a two-parameter Weibull distribution. Use of Weibull functions with overall survival significantly increases the precision of small arms typical of early phase trials: analysis of a 50-patient trial arm using parametric forms is as precise as traditional, non-parametric analysis of a 90-patient arm. We also show that frequent deviations from the Cox proportional hazards assumption, particularly in trials of immune checkpoint inhibitors, arise from time-dependent therapeutic effects. Trial duration therefore has an underappreciated impact on the likelihood of success.
- Published
- 2021
27. Divining responder populations from survival data
- Author
-
Alyssa M. Vanderbeek, Brian M. Alexander, Steffen Ventz, Lorenzo Trippa, Geoffrey Fell, and Rifaquat Rahman
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Treatment response ,medicine.medical_treatment ,Population ,03 medical and health sciences ,0302 clinical medicine ,Survival data ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Humans ,Precision Medicine ,education ,Survival analysis ,Retrospective Studies ,education.field_of_study ,Models, Statistical ,Brain Neoplasms ,business.industry ,Chemoradiotherapy ,Glioma ,Hematology ,Prognosis ,Precision medicine ,Survival Rate ,Clinical trial ,Radiation therapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,business ,Follow-Up Studies - Abstract
Background Biomarkers that predict treatment response are the foundation of precision medicine in clinical decision-making and have the potential to significantly improve the efficiency of clinical trials. Such biomarkers may be identified before clinical testing but many trials enroll unselected populations. We hypothesized that time-varying treatment effects in unselected trials may result from identifiable responder subpopulations that may have associated biomarkers. Materials and methods We first simulated scenarios of clinical trials with biomarker populations of varying prevalence and prognostic and predictive associations to illustrate the impact of subgroup-specific effects on overall population estimates. To show a real-world example of time-dependent treatment effects resulting from a prognostic and predictive biomarker, we re-analyzed data from a published clinical trial (RTOG, Radiation Therapy Oncology Group, 9402). We then demonstrated a quantitative framework to fit survival data from clinical trials using statistical models incorporating known estimates of biomarker prevalence and prognostic value to prioritize predictive biomarker hypotheses. Results Our simulation studies demonstrate how biomarker subgroups that are both predictive and prognostic can manifest as time-dependent treatment effects in overall populations. RTOG 9402 provides a representative example where 1p/19q co-deletion and IDH mutation biomarker-specific effects led to time-varying treatment effects and a considerable deviation from proportional hazards in the overall trial population. Finally, using biomarker data from The Cancer Genome Atlas, we were able to generate statistical models that correctly identified and prioritized a commonly used biomarker through retrospective analysis of published clinical trial data. Conclusions Biomarkers that are both predictive and prognostic can result in characteristic changes in survival results. Retrospectively analyzing survival data from clinical trials may highlight potential indications for which an underlying predictive biomarker may be found.
- Published
- 2019
28. Phase 2 study of response-guided neoadjuvant sacituzumab govitecan (IMMU-132) in patients with localized triple-negative breast cancer: Results from the NeoSTAR trial
- Author
-
Laura Spring, Sara M. Tolaney, Neelam Vijay Desai, Geoffrey Fell, Lorenzo Trippa, Amy H. Comander, Therese Marie Mulvey, Shannon McLaughlin, Phoebe Ryan, Aron S. Rosenstock, Ana Christina Garrido-Castro, Filipa Lynce, Beverly Moy, Steven J. Isakoff, Nadine M. Tung, Elizabeth A. Mittendorf, Leif W. Ellisen, and Aditya Bardia
- Subjects
Cancer Research ,Oncology - Abstract
512 Background: Sacituzumab govitecan (SG), a novel antibody-drug conjugate in which the topoisomerase 1 inhibitor SN-38 (active metabolite of irinotecan) is linked to a humanized monoclonal antibody targeting the tumor antigen Trop2, is currently approved for treatment of patients (pts) with pre-treated metastatic triple negative breast cancer (TNBC). We conducted a phase 2 study evaluating neoadjuvant (NA) SG as upfront therapy for pts with localized TNBC (NCT04230109). The primary objective was to assess pathological complete response (pCR) rate in breast and lymph nodes (ypT0/isN0) with SG. Secondary objectives included assessment of radiological response rate, evaluation of the safety and tolerability (CTCAE v5.0) and event-free survival (EFS). Methods: Patients with localized TNBC (tumor size ≥1cm, or any size if node positive) with no prior treatment were eligible. SG was administered IV on Days 1, 8 of each 21-day cycle at a starting dose of 10 mg/kg for 4 cycles. After 4 cycles, patients with biopsy-proven residual disease, considered as no pCR for primary endpoint, had the option to receive additional NA therapy at the discretion of the treating physician. Radiologic response (US or MRI) was defined by RECIST version 1.1 using a composite response of CR & PR. Standard descriptive statistics were utilized, including 95% binomial confidence intervals for all rates estimated. Results: From 7/14/20 – 8/31/21, 50 pts were enrolled (median age = 48.5; 11 stage I disease, 24 stage II, 11 stage III, 4 unknown; 62% node negative). The majority (98%; n = 49) of pts completed 4 cycles of SG. Overall, the radiological response rate with SG alone was 62% (n = 31, 95% CI 48%, 77%). 26 pts proceeded directly to surgery after SG. Overall, the pCR rate with SG alone was 30% (n = 15/50, 95% CI 18%, 45%). The other 11 pts had RCB-1 (n = 3), RCB-2 (n = 5), and RCB-3 (n = 3) disease, respectively. Of the 24 pts who received additional NA therapy, 6 had a pCR (3 received anthracycline-based regimen, 2 carboplatin/taxane, and 1 docetaxel/cyclophosphamide). Among pts with a germline BRCA mutation (n = 8), 7 proceeded directly to surgery after SG and 6 had a pCR (86%, 95% CI 42%, 99%). The most common AEs with SG were nausea (82%, n = 41), fatigue (78%, n = 39), alopecia (76%, n = 38), neutropenia (58%, n = 29), anemia (36%, n = 18), and rash (48%, n = 24). 6% of pts required dose-reduction. No pts discontinued SG therapy due to disease progression or AEs; 1 discontinued due to minimal response per investigator preference. At the time of data cut-off (1/18/22), no pts experienced disease recurrence. Updated biomarker and EFS results will be presented at the meeting. Conclusions: In the first neoadjuvant trial in TNBC with an ADC, SG demonstrated single agent efficacy in localized TNBC. Further research on optimal duration of SG as well as NA combination strategies, including immunotherapy, are needed. Clinical trial information: NCT04230109.
- Published
- 2022
29. Cancer patient survival can be accurately parameterized, revealing time-dependent therapeutic effects and doubling the precision of small trials
- Author
-
Peter K. Sorger, Deborah Plana, Brian M. Alexander, Adam C. Palmer, and Geoffrey Fell
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Therapeutic effect ,Hazard ratio ,Parameterized complexity ,Cancer ,Patient survival ,medicine.disease ,Clinical trial ,Internal medicine ,medicine ,business ,Weibull distribution ,Parametric statistics - Abstract
SUMMARYIndividual participant data (IPD) from completed oncology clinical trials are a valuable but rarely available source of information. A lack of minable survival distributions has made it difficult to identify factors determining the success and failure of clinical trials and improve trial design. We imputed survival IPD from ∼500 arms of phase III oncology trials (representing ∼220,000 events) and found that they are well fit by a two-parameter Weibull distribution. This makes it possible to use parametric statistics to substantially increase trial precision with small patient cohorts typical of phase I or II trials. For example, a 50-person trial parameterized using Weibull distributions is as precise as a 90-person trial evaluated using traditional statistics. Mining IPD also showed that frequent violations of the proportional hazards assumption, particularly in trials of immune checkpoint inhibitors (ICIs), arise from time-dependent therapeutic effects and hazard ratios. Thus, the duration of ICI trials has an underappreciated impact on the likelihood of their success.
- Published
- 2021
30. Assessment of Simulated SARS-CoV-2 Infection and Mortality Risk Associated With Radiation Therapy Among Patients in 8 Randomized Clinical Trials
- Author
-
Shyam K. Tanguturi, Steffen Ventz, Jennifer R. Bellon, Shervin Tabrizi, Anthony V. D'Amico, Rifaquat Rahman, Daphne A. Haas-Kogan, Ayal A. Aizer, Brian M. Alexander, Geoffrey Fell, Lorenzo Trippa, Harvey J. Mamon, Daniel N. Cagney, and Paul L. Nguyen
- Subjects
Male ,Risk ,Oncology ,Comparative Effectiveness Research ,medicine.medical_specialty ,Datasets as Topic ,Breast Neoplasms ,Risk Assessment ,law.invention ,Breast cancer ,Randomized controlled trial ,law ,Internal medicine ,Case fatality rate ,medicine ,Humans ,Pandemics ,Proportional Hazards Models ,Original Investigation ,Randomized Controlled Trials as Topic ,Infection Control ,Rectal Neoplasms ,Proportional hazards model ,business.industry ,SARS-CoV-2 ,Mortality rate ,Research ,Dose fractionation ,Prostatic Neoplasms ,COVID-19 ,Standard of Care ,General Medicine ,medicine.disease ,Clinical trial ,Online Only ,Female ,Radiation Dose Hypofractionation ,Dose Fractionation, Radiation ,Patient Care ,Radiology ,Risk assessment ,business ,Algorithms - Abstract
Key Points Question How is the COVID-19 pandemic associated with risks and benefits of standard cancer therapy? Findings In this comparative effectiveness study reanalyzing data from 8 randomized trials across oncology, radiation fractionation was not associated with outcomes in low COVID-19–risk scenarios. In higher-risk scenarios, aggressive hypofractionation was associated with a survival benefit, whereas moderate hypofractionation was not. Meaning These findings suggest that the expected benefit of altering radiation therapy during the COVID-19 pandemic is associated with local pandemic factors and the specifics of treatment modification, and can be estimated using a quantitative framework based on completed randomized trials to support treatment decision-making., This comparative effectiveness study investigates how the COVID-19 pandemic is associated with the benefits and risks of standard radiation therapy in simulated patients., Importance During the COVID-19 pandemic, cancer therapy may put patients at risk of SARS-CoV-2 infection and mortality. The impacts of proposed alternatives on reducing infection risk are unknown. Objective To investigate how the COVID-19 pandemic is associated with the risks and benefits of standard radiation therapy (RT). Design, Setting, and Participants This comparative effectiveness study used estimated individual patient–level data extracted from published Kaplan-Meier survival figures from 8 randomized clinical trials across oncology from 1993 to 2014 that evaluated the inclusion of RT or compared different RT fractionation regimens. Included trials were Dutch TME and TROG 01.04 examining rectal cancer; CALGB 9343, OCOG hypofractionation trial, FAST-Forward, and NSABP B-39 examining early stage breast cancer, and CHHiP and HYPO-RT-PC examining prostate cancer. Risk of SARS-CoV-2 infection and mortality associated with receipt of RT in the treatment arms were simulated and trials were reanalyzed. Data were analyzed between April 1, 2020, and June 30, 2020. Exposures COVID-19 risk associated with treatment was simulated across different pandemic scenarios, varying infection risk per fractions (IRFs) and case fatality rates (CFRs). Main Outcomes and Measures Overall survival was evaluated using Cox proportional hazards modeling under different pandemic scenarios. Results Estimated IPLD from a total of 14 170 patients were included in the simulations. In scenarios with low COVID-19-associated risks (IRF, 0.5%; CFR, 5%), fractionation was not significantly associated with outcomes. In locally advanced rectal cancer, short-course RT was associated with better outcomes than long-course chemoradiation (TROG 01.04) and was associated with similar outcomes as RT omission (Dutch TME) in most settings (eg, TROG 01.04 median HR, 0.66 [95% CI, 0.46-0.96]; Dutch TME median HR, 0.91 [95% CI, 0.80-1.03] in a scenario with IRF 5% and CFR 20%). Moderate hypofractionation in early stage breast cancer (OCOG hypofractionation trial) and prostate cancer (CHHiP) was not associated with survival benefits in the setting of COVID-19 (eg, OCOG hypofractionation trial median HR, 0.89 [95% CI, 0.74-1.06]; CHHiP median HR, 0.87 [95% CI, 0.75-1.01] under high-risk scenario with IRF 10% and CFR 30%). More aggressive hypofractionation (FAST-Forward, HYPO-RT-PC) and accelerated partial breast irradiation (NSABP B-39) were associated with improved survival in higher risk scenarios (eg, FAST-Forward median HR, 0.58 [95% CI, 0.49-0.68]; HYPO-RT-PC median HR, 0.60 [95% CI, 0.48-0.75] under scenario with IRF 10% and CFR 30%). Conclusions and Relevance In this comparative effectiveness study of data from 8 clinical trials of patients receiving radiation therapy to simulate COVID-19 risk and mortality rates, treatment modification was not associated with altered risk from COVID-19 in lower-risk scenarios and was only associated with decreased mortality in very high COVID-19–risk scenarios. This model, which can be adapted to dynamic changes in COVID-19 risk, provides a flexible, quantitative approach to assess the potential impact of treatment modifications and supports the continued delivery of standard evidence-based care with appropriate precautions against COVID-19.
- Published
- 2021
31. P-089: Identification of novel targets in multiple myeloma for 'undruggable' RAS/CDK signaling cascade
- Author
-
Yu-Tzu Tai, Kenneth C. Anderson, Kenneth Wen, Ivane Abiatari, David M. Dorfman, Sophia Adamia, Morgan OKeefe, Shruti Bhatt, Anthony Letai, Geoffrey Fell, Zuzana Chyra, and Teru Hideshima
- Subjects
Cancer Research ,Spliceosome ,Gene knockdown ,biology ,business.industry ,Intron ,Hematology ,Oncology ,Downregulation and upregulation ,Cyclin-dependent kinase ,RNA splicing ,Cancer research ,biology.protein ,Medicine ,Epigenetics ,business ,Gene - Abstract
Background RAS/CDK-dependent pathways play essential roles in multiple myeloma (MM) pathogenesis; both pathways are undruggable. We evaluated molecular changes associated with pathway-level responses after RAS/CDK inhibition to identify novel molecular targets. Method in our previous studies MM cells were treated with selected Erk1/2 and CDK4/6 inhibitors (Ei, Ci) to target RAS/CDK pathways. Our studies indicate strong synergistic (IC Results We evaluated mRNA splicing changes in MM cells, with and without Erk1/2 knockdown or with Ei+Ci treatment. Unsupervised clustering of deregulated genes showed dose-dependent treatment effects. Upregulation in response to Erk1/2 knockdown and downregulation due to treatment with Ei+Ci were considered spliced gene signatures linked to RAS/CDK modulation. Gene/pathway enrichment analyses of these genes showed their involvement in cell proliferation and regulation of epigenetic networks in MM. Importantly, these analyses suggest that overexpression of RAVER1/SNRPB core splicing regulator genes are associated with RAS/CDK pathway regulation. These genes encode subunits of U1/2/4/5 spliceosome complexes and are involved in intron retention processes, a marker of malignant transformation. We evaluated expressions of RAVER1 and SNRPB in 558 MM patent samples and 10 normal donor BM PCs and observed significant (p Conclusions Our studies show an association between RNA processing and RAS-CDK pathways in MM, identify a core splicing protein, SNRPB, as a novel target for modulating this undruggable cascade, and suggest that targeting spliceosome complexes is a promising therapy.
- Published
- 2021
32. KMDATA: a curated database of reconstructed individual patient-level data from 153 oncology clinical trials
- Author
-
Jon McDunn, Bill Louv, Lorenzo Trippa, Brian M. Alexander, Geoffrey Fell, Robert A. Redd, Rifaquat Rahman, Andrea Arfè, Alyssa M Vanderbeek, and Steffen Ventz
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Databases, Factual ,MEDLINE ,Kaplan-Meier Estimate ,Medical Oncology ,computer.software_genre ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Internal medicine ,medicine ,Overall survival ,Humans ,030212 general & internal medicine ,Survival analysis ,Database ,Extramural ,business.industry ,Clinical trial ,Patient level data ,030220 oncology & carcinogenesis ,AcademicSubjects/SCI00960 ,Original Article ,General Agricultural and Biological Sciences ,business ,computer ,Information Systems ,Type I and type II errors - Abstract
We created a database of reconstructed patient-level data from published clinical trials that includes multiple time-to-event outcomes such as overall survival and progression-free survival. Outcomes were extracted from Kaplan–Meier (KM) curves reported in 153 oncology Phase III clinical trial publications identified through a PubMed search of clinical trials in breast, lung, prostate and colorectal cancer, published between 2014 and 2016. For each trial that met our search criteria, we curated study-level information and digitized all reported KM curves with the software Digitizelt. We then used the digitized KM survival curves to estimate (possibly censored) patient-level time-to-event outcomes. Collections of time-to-event datasets from completed trials can be used to support the choice of appropriate trial designs for future clinical studies. Patient-level data allow investigators to tailor clinical trial designs to diseases and classes of treatments. Patient-level data also allow investigators to estimate the operating characteristics (e.g. power and type I error rate) of candidate statistical designs and methods. Database URL: https://10.6084/m9.figshare.14642247.v1
- Published
- 2021
33. Metabolic perturbations sensitize triple-negative breast cancers to apoptosis induced by BH3 mimetics
- Author
-
David T. Scadden, Kristopher A. Sarosiek, Isaac S. Harris, Joan S. Brugge, Vinícius Guimarães Ferreira, Danielle S. Potter, Veerle W. Daniels, Salma Parvin, Geoffrey Fell, Jennifer E. Endress, Patrick Bhola, Jason J. Zoeller, Rajat M. Gupta, Johan Spetz, Nick van Gastel, Emanuel Carrilho, Kelley E. McQueeney, Anthony Letai, and Binyam Yilma
- Subjects
Nicotinamide phosphoribosyltransferase ,Priming (immunology) ,Apoptosis ,Triple Negative Breast Neoplasms ,Biochemistry ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Humans ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Cell Biology ,Mitochondria ,Enzyme ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,NAD+ kinase ,METABOLISMO ,Signal transduction ,Apoptosis Regulatory Proteins - Abstract
Cancer cells have differential metabolic dependencies compared to their non-malignant counterparts. However, few metabolism-targeting compounds have been successful in clinical trials. Here, we investigated the metabolic vulnerabilities of triple-negative breast cancer (TNBC), in particular those metabolic perturbations that increased mitochondrial apoptotic priming and sensitivity to BH3 mimetics (drugs that antagonize anti-apoptotic proteins). We used high throughput-dynamic BH3 profiling (HT-DBP) to screen a library of metabolism-perturbing small molecules, which revealed inhibitors of the enzyme nicotinamide phosphoribosyltransferase (NAMPT) as top candidates. In some TNBC cells but not in non-malignant cells, NAMPT inhibitors increased overall apoptotic priming and induced dependencies on specific anti-apoptotic BCL-2 family members. Treatment of TNBC cells with NAMPT inhibitors sensitized them to subsequent treatment with BH3 mimetics. The combination of a NAMPT inhibitor (FK866) and an MCL-1 antagonist (S63845) reduced tumor growth in a TNBC patient-derived xenograft model in vivo. We found that NAMPT inhibition reduced NAD(+) concentrations below a critical threshold that resulted in depletion of adenine, which was the metabolic trigger that primed TNBC cells for apoptosis. These findings demonstrate a close interaction between metabolic and mitochondrial apoptotic signaling pathways and reveal that exploitation of a tumor-specific metabolic vulnerability can sensitize some TNBC to BH3 mimetics.
- Published
- 2021
34. Clinical Significance of CBC and WBC Morphology in the Diagnosis and Clinical Course of COVID-19 Infection
- Author
-
Jean M. Connors, Geoffrey Fell, Elisabeth M. Battinelli, Olga Pozdnyakova, Nathan T. Connell, and Annette S. Kim
- Subjects
Morphology ,Male ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Neutrophils ,Critical Illness ,Peripheral blood ,Vacuolization ,medicine.disease_cause ,Virus ,Monocytes ,law.invention ,Leukocyte Count ,law ,Internal medicine ,Lymphopenia ,medicine ,Leukocytes ,CBC research parameters ,Humans ,Clinical significance ,Coronavirus ,Aged ,Atypical Lymphocyte ,medicine.diagnostic_test ,business.industry ,SARS-CoV-2 ,Complete blood count ,COVID-19 ,General Medicine ,Middle Aged ,Intensive care unit ,Neutrophilia ,Blood Cell Count ,Disease Progression ,Original Article ,Female ,medicine.symptom ,Atypical lymphocytes ,business ,AcademicSubjects/MED00690 - Abstract
Objectives To investigate the clinical significance of numeric and morphologic peripheral blood (PB) changes in coronavirus disease 2019 (COVID-19)–positive patients in predicting the outcome, as well as to compare these changes between critically ill COVID-19–positive and COVID-19–negative patients. Methods The study included 90 COVID-19–positive (51 intensive care unit [ICU] and 39 non-ICU) patients and 30 COVID-19–negative ICU patients. We collected CBC parameters (both standard and research) and PB morphologic findings, which were independently scored by two hematopathologists. Results All patients with COVID-19 demonstrated striking numeric and morphologic WBC changes, which were different between mild and severe disease states. More severe disease was associated with significant neutrophilia and lymphopenia, which was intensified in critically ill patients. Abnormal WBC morphology, most pronounced in monocytes and lymphocytes, was associated with more mild disease; the changes were lost with disease progression. Between COVID-19–positive and COVID-19–negative ICU patients, significant differences in morphology-associated research parameters were indicative of changes due to the severe acute respiratory syndrome coronavirus 2 virus, including higher RNA content in monocytes, lower RNA content in lymphocytes, and smaller hypogranular neutrophils. Conclusions Hospitalized patients with COVID-19 should undergo a comprehensive daily CBC with manual WBC differential to monitor for numerical and morphologic changes predictive of poor outcome and signs of disease progression.
- Published
- 2020
- Full Text
- View/download PDF
35. Reply to 'Keynote 48: Is it really for everyone?'
- Author
-
Lorenzo Trippa, Jonathan D. Schoenfeld, Geoffrey Fell, and Robert I. Haddad
- Subjects
Cancer Research ,Oncology ,Philosophy ,MEDLINE ,Library science ,Oral Surgery - Published
- 2020
36. CTNI-11. CC-115 IN NEWLY DIAGNOSED MGMT UNMETHYLATED GLIOBLASTOMA IN THE INDIVIDUALIZED SCREENING TRIAL OF INNOVATIVE GLIOBLASTOMA THERAPY (INSIGHT): A PHASE II RANDOMIZED BAYESIAN ADAPTIVE PLATFORM TRIAL
- Author
-
Lorenzo Trippa, David Meredith, E. Antonio Chiocca, Mehdi Touat, Lakshmi Nayak, Mary Welch, David Schiff, Jack Geduldig, Omar Arnaout, Louis B. Nabors, Christine McCluskey, Rameen Beroukhim, Thomas Kaley, Mikael L. Rinne, Sandro Santagata, Howard Colman, Shyam K. Tanguturi, Manmeet Ahluwalia, Brian M. Alexander, Rifaquat Rahman, Daniel N. Cagney, Andrew B. Lassman, David A. Reardon, Patrick Y. Wen, Maria Lavallee, Geoffrey Fell, Ugonma Chukwueke, Heinrich Elinzano, Ayal A. Aizer, Keith L. Ligon, Jose Mcfaline-Figueroa, Fiona Watkinson, Isabel Arrillaga-Romany, Jaroslaw T. Hepel, Daphne A. Haas-Kogan, Lisa Doherty, Christine Lu-Emerson, Tracy T. Batchelor, Shanna Dowling, Wenya Linda Bi, Jennifer Brunno, Evanthia Galanis, Brittany Fisher-Longden, Jan Drappatz, Sarah C. Gaffey, and Eudocia Q. Lee
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,Screening Trial ,medicine.medical_treatment ,Clinical Trials: Non-Immunologic ,O-6-methylguanine-DNA methyltransferase ,medicine.disease ,Radiation therapy ,Internal medicine ,medicine ,MGMT-Unmethylated Glioblastoma ,Neurology (clinical) ,Liver function ,Progression-free survival ,business ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND CC-115 is an oral, CNS-penetrant, selective inhibitor of mammalian target of rapamycin kinase (mTOR) and deoxyribonucleic acid-dependent protein kinase (DNA-PK). Both targets are important in glioblastoma; PI3K/Akt/mTOR signaling is hyperactive in most glioblastomas, and DNA-PK is integral to repair of radiotherapy-mediated DNA damage. To investigate CC-115 in newly diagnosed glioblastoma and explore potential genomic biomarker associations, CC-115 was evaluated in the Individualized Screening Trial of Innovative Glioblastoma Therapy (INSIGhT) trial, an adaptive platform trial designed to efficiently test experimental agents. METHODS Adults with newly diagnosed MGMT-unmethylated glioblastoma, with genomic data available, are eligible for this ongoing trial. Patients are adaptively randomized to one of several experimental arms or the control arm: standard radiotherapy with concurrent and adjuvant temozolomide. The primary endpoint is overall survival (OS). Patients randomized to CC-115 (10mg po BID) received it concurrently with radiotherapy and as adjuvant monotherapy. As the first in-human use of CC-115 with radiation, a safety lead-in 3 + 3 design was used. RESULTS Twelve patients were randomized to CC-115; seven patients had possible treatment-related CTCAE grade > 3 toxicity, including four pre-specified dose-limiting toxicities: liver function abnormality (n=1), hyperlipidemia (n=1), lipase elevation (n=1) and cerebral edema (n=1). There was no significant difference in progression-free survival (PFS, median 4.2 months [CC-115] vs. 5.2 months, p=0.9) or OS (median 10.1 months [CC-115] vs. 14.5 months, p=0.9) compared to the 50 patients randomized to the control arm. Based on early PFS results, randomization probability to CC-115 decreased from 25% to < 10% at time of the trial arm closure. CONCLUSION Concurrent and adjuvant CC-115 was associated with toxicity and failed to improve PFS or OS. The INSIGhT trial design allowed for more efficient testing of CC-115, decreasing patients and resources allocated to a therapy that was discontinued due to concerns about toxicity and unfavorable risk-to-benefit ratio.
- Published
- 2020
37. The adjuvant use of capecitabine for residual disease following pre-operative chemotherapy for breast cancer: Challenges applying CREATE-X to a US population
- Author
-
Michelle C. Specht, Geoffrey Fell, Rachel B. Jimenez, Therese M. Mulvey, Beverly Moy, Andrew D. Johnson, Kassidy Beyerlin, Aditya Bardia, Christine E Edmonds, Steven J. Isakoff, Mark L. Zangardi, Veerle Bossuyt, Leif W. Ellisen, and Laura Spring
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Population ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,Disease ,Disease-Free Survival ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Pre-operative chemotherapy ,Medicine ,Humans ,Pharmacology (medical) ,030212 general & internal medicine ,education ,Triple-negative breast cancer ,Neoadjuvant therapy ,Retrospective Studies ,education.field_of_study ,business.industry ,medicine.disease ,Neoadjuvant Therapy ,Treatment Outcome ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Female ,Fluorouracil ,business ,Adjuvant ,medicine.drug - Abstract
Introduction The CREATE-X study, conducted in Japan and South Korea, established capecitabine as an adjuvant treatment option for patients with triple negative breast cancer (TNBC) who have residual disease (RD) following neoadjuvant anthracycline or taxane-based chemotherapy. However, there are no reports on the tolerability and outcomes of adjuvant capecitabine in the US setting following publication of the CREATE-X data. Methods We retrospectively collected treatment and tolerability data from the medical records of the first 23 TNBC patients who received adjuvant capecitabine for RD post neoadjuvant chemotherapy at our institution. Disease-free survival was assessed using the Kaplan-Meier method. Results The median starting dosage of capecitabine was 1871 mg/m2/day, most commonly divided into two daily doses on days 1-14 of each 21 day cycle. 34.8% of patients completed the treatment as prescribed. Side effects associated with treatment were common with 69.6% of patients experiencing hand-foot syndrome, 39.1% of patients experiencing diarrhea, and 13.0% of patients requiring hospitalization for side effects. Of 23 patients treated with adjuvant capecitabine, 34.8% completed the planned dose, 30.4% completed with dose reduction, and 34.8% discontinued early. At a median follow-up time of 14 months, the median disease-free survival was 22 months, with 30.4% of patients experiencing recurrence. Conclusion Tolerability was poor overall compared to the CREATE-X cohort. Administering adjuvant capecitabine for TNBC patients with residual disease in the United States is challenging given differences in tolerability. More research is needed to understand how poor tolerability will affect the efficacy of this approach in the US population.
- Published
- 2020
38. Keynote 48: Is it really for everyone?
- Author
-
Jonathan D. Schoenfeld, Robert I. Haddad, Lorenzo Trippa, and Geoffrey Fell
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,Cetuximab ,business.industry ,First line ,medicine.medical_treatment ,Pembrolizumab ,medicine.disease ,Head and neck squamous-cell carcinoma ,Survival benefit ,Internal medicine ,medicine ,Single agent ,In patient ,business ,medicine.drug - Abstract
Burtness et al. recently published the landmark Keynote-48 study, demonstrating a survival benefit for pembrolizumab monotherapy and pembrolizumab/chemotherapy, compared with cetuximab/chemotherapy, in patients with recurrent/metastatic head and neck squamous cell carcinoma (HNSCC). These data are impactful and practice-changing, and have rapidly been adopted in practice, with increasing numbers of HNSCC patients receiving either pembrolizumab monotherapy, or pembrolizumab / chemotherapy, in the first line recurrent / metastatic setting. Pembrolizumab was approved as a single agent for patients whose tumors express PD□L1 (Combined Positive Score [CPS] ≥1), while pembrolizumab / chemotherapy was approved for use in the United States for all patients irrespective of PD-L1 expression.
- Published
- 2020
- Full Text
- View/download PDF
39. Pathological complete response after neoadjuvant chemotherapy and impact on breast cancer recurrence and survival: a comprehensive meta-analysis
- Author
-
Brian M. Alexander, Lorenzo Trippa, Barbara L. Smith, Andrea Arfè, Laura Spring, Rachel A. Greenup, Beverly Moy, Aditya Bardia, Kerry L. Reynolds, Chandni Sharma, Steven J. Isakoff, Geoffrey Fell, and Giovanni Parmigiani
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Breast Neoplasms ,Disease ,Disease-Free Survival ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Adjuvant therapy ,Humans ,Neoadjuvant therapy ,Chemotherapy ,business.industry ,Bayes Theorem ,medicine.disease ,Prognosis ,Neoadjuvant Therapy ,030104 developmental biology ,Nat ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Meta-analysis ,Neoplasm Recurrence, Local ,business ,Adjuvant - Abstract
Purpose:While various studies have highlighted the prognostic significance of pathologic complete response (pCR) after neoadjuvant chemotherapy (NAT), the impact of additional adjuvant therapy after pCR is not known.Experimental Design:PubMed was searched for studies with NAT for breast cancer and individual patient-level data was extracted for analysis using plot digitizer software. HRs, with 95% probability intervals (PI), measuring the association between pCR and overall survival (OS) or event-free survival (EFS), were estimated using Bayesian piece-wise exponential proportional hazards hierarchical models including pCR as predictor.Results:Overall, 52 of 3,209 publications met inclusion criteria, totaling 27,895 patients. Patients with a pCR after NAT had significantly better EFS (HR = 0.31; 95% PI, 0.24–0.39), particularly for triple-negative (HR = 0.18; 95% PI, 0.10–0.31) and HER2+ (HR = 0.32; 95% PI, 0.21–0.47) disease. Similarly, pCR after NAT was also associated with improved survival (HR = 0.22; 95% PI, 0.15–0.30). The association of pCR with improved EFS was similar among patients who received subsequent adjuvant chemotherapy (HR = 0.36; 95% PI, 0.19–0.67) and those without adjuvant chemotherapy (HR = 0.36; 95% PI, 0.27–0.54), with no significant difference between the two groups (P = 0.60).Conclusions:Achieving pCR following NAT is associated with significantly better EFS and OS, particularly for triple-negative and HER2+ breast cancer. The similar outcomes with or without adjuvant chemotherapy in patients who attain pCR likely reflects tumor biology and systemic clearance of micrometastatic disease, highlighting the potential of escalation/deescalation strategies in the adjuvant setting based on neoadjuvant response.See related commentary by Esserman, p. 2771
- Published
- 2020
40. Endogenous Glucocorticoid Signaling Regulates CD8+ T Cell Differentiation and Development of Dysfunction in the Tumor Microenvironment
- Author
-
Giulia Escobar, Nandini Acharya, Meromit Singer, Huiyuan Zhang, Marcus Bosenberg, Vijay K. Kuchroo, Karen O. Dixon, Davide Mangani, Aviv Regev, Geoffrey Fell, Elena Christian, Junrong Xia, Donna Neuberg, Orit Rozenblatt-Rosen, Michelle Ferreira, Asaf Madi, Shai Dulberg, Katherine Tooley, Max Klapholz, and Ana C. Anderson
- Subjects
0301 basic medicine ,Tumor microenvironment ,medicine.medical_treatment ,Immunology ,Biology ,03 medical and health sciences ,Steroid hormone ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Glucocorticoid receptor ,Cancer immunotherapy ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Immunology and Allergy ,Cytotoxic T cell ,Receptor ,Transcription factor ,Glucocorticoid ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Summary Identifying signals in the tumor microenvironment (TME) that shape CD8+ T cell phenotype can inform novel therapeutic approaches for cancer. Here, we identified a gradient of increasing glucocorticoid receptor (GR) expression and signaling from naive to dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). Conditional deletion of the GR in CD8+ TILs improved effector differentiation, reduced expression of the transcription factor TCF-1, and inhibited the dysfunctional phenotype, culminating in tumor growth inhibition. GR signaling transactivated the expression of multiple checkpoint receptors and promoted the induction of dysfunction-associated genes upon T cell activation. In the TME, monocyte-macrophage lineage cells produced glucocorticoids and genetic ablation of steroidogenesis in these cells as well as localized pharmacologic inhibition of glucocorticoid biosynthesis improved tumor growth control. Active glucocorticoid signaling associated with failure to respond to checkpoint blockade in both preclinical models and melanoma patients. Thus, endogenous steroid hormone signaling in CD8+ TILs promotes dysfunction, with important implications for cancer immunotherapy.
- Published
- 2020
41. Identification of Novel Targets Based on Splicing Alterations for Undruggable RAS/CDK Signaling Cascade in Multiple Myeloma
- Author
-
Yu-Tzu Tai, Shruti Bhatt, Sophia Adamia, Kenneth Wen, Kenneth C. Anderson, Teru Hideshima, David M. Dorfman, Anthony Letai, Zuzana Chyra, Ivane Abiatari, Morgan O'Keefe, and Geoffrey Fell
- Subjects
biology ,Immunology ,Cell Biology ,Hematology ,Computational biology ,medicine.disease ,Biochemistry ,Cascade ,Cyclin-dependent kinase ,RNA splicing ,medicine ,biology.protein ,Identification (biology) ,Multiple myeloma - Abstract
Background: RAS/CDK-dependent pathways play essential roles in multiple myeloma (MM) pathogenesis. Targeting these pathways represents a novel therapeutic strategy in MM. Our ongoing studies (>420 patients) demonstrate that aberrantly spliced transcript expressions can predict MM patient survival outcomes better than gene expression alone, indicating a significant role of splicing mechanism in MM pathophysiology. These studies also identified intron retentions as the predominant recurrent alterations (~32% of spliced genes were retained introns) in MM. We evaluated splicing alterations associated with pathway-level responses after RAS/CDK inhibition in order to identify and validate novel molecular targets. Methods/results: MM cells were treated with selected Erk1/2 and CDK4/6 inhibitors (Ei, Ci) to inhibit RAS and CDK pathways. Our studies demonstrated strong synergistic (IC We next evaluated aberrantly spliced transcript expression in MM cells, with/without Erk1/2 knockdown (KD) or with Ei+Ci treatment. Unsupervised clustering of deregulated genes showed dose-dependent treatment effects. This observation was further supported by principal component analyses: upregulation in response to Erk1/2 KD and downregulation due to treatment with Ei+Ci were considered spliced gene-signatures linked to RAS/CDK modulation. Gene/pathway enrichment analyses of these genes showed their involvement in cell proliferation and regulation of epigenetic networks in MM. Importantly, these analyses suggest that overexpression of RAVER1/SNRPB core splicing regulator genes are associated with RAS/CDK pathway regulation. These genes encode subunits of U1/2/4/5 spliceosome complex and are involved in intron retention processes, a marker of malignant transformation. We compared signature-gene expressions from 558 MM patient samples to the signature-genes in plasma cells from normal donors and observed significant (p Conclusions: Our studies 1) show an association between RNA processing and RAS-CDK pathways in MM, 2) identify a core splicing protein, SNRPB/RAVER1, as a novel target for modulating this cascade, and 3) suggest that targeting spliceosome complexes represents a promising therapy in MM. Disclosures Letai: Zentalis Pharmaceuticals: Other: equity holding member of the scientific advisory board; Dialectic Therapeutics: Other: equity holding member of the scientific advisory board; Flash Therapeutics: Other: equity holding member of the scientific advisory board. Anderson: Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Scientific Founder of Oncopep and C4 Therapeutics: Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company; AstraZeneca: Membership on an entity's Board of Directors or advisory committees; Mana Therapeutics: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
42. Maximal Tolerated Dose of the BCL-2 Inhibitor Venetoclax in Combination with Daunorubicin/Cytarabine Induction in Previously Untreated Adults with Acute Myeloid Leukemia (AML)
- Author
-
Marina Konopleva, Francis Brown, Daniel J. DeAngelo, Jacqueline S. Garcia, Yael Flammand, Marlise R. Luskin, Geoffrey Fell, Anthony Letai, Michael McGinnis, Jeremy M. Stewart, and Richard Stone
- Subjects
medicine.medical_specialty ,business.industry ,Venetoclax ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,Biochemistry ,Bcl-2 Inhibitor ,Regimen ,chemistry.chemical_compound ,chemistry ,Younger adults ,Maximum tolerated dose ,Internal medicine ,Ven ,Cytarabine ,Medicine ,business ,medicine.drug - Abstract
Background: Although daunorubicin/cytarabine ("3+7")-based chemotherapy produces complete remission (CR) rates of 70% in younger adults, a significant proportion require re-induction to achieve CR or are refractory. Among those achieving CR, relapse after post-remission therapy is common and limits cure. Venetoclax (VEN) is an oral, selective BCL-2 inhibitor that promotes mitochondrial-mediated apoptosis in myeloblasts during cytotoxic stress. VEN plus hypomethylating agents in untreated unfit elderly AML patients (pts) elicits high response rates and improves overall survival. We hypothesize that VEN in combination with "3+7" could lead to a higher rate of measurable residual disease (MRD)-negative CR, perhaps obviating the need for reinduction as well as limiting relapse without undue toxicity. Methods: The primary objective of this phase 1 study was to determine the maximum tolerated dose (MTD) of VEN plus "3+7" induction and cytarabine-based consolidation therapy in previously untreated AML pts. Pt eligibility includes previously untreated AML, Results: Two of the first 6 pts on VEN 200 experienced DLTs (rapidly resolving grade 4 DIC after a single dose of VEN 50 mg and death at day 14 of sepsis in a 73 year old man). Given 2/6 DLT events, an amendment allowed further accrual at the starting dose of VEN 200 mg but restricted accrual to age 18-60 years. All 3 pts on the expanded cohort and 3/3 at the 400 mg cohort have responded, and none experienced a DLT. The trial expanded to treat pts with 600 mg of VEN, but a 24 yo F experienced a septic death on day 9. Therefore, the 400 mg dose level was determined to be the MTD. At the VEN 200 dose level (n=7), the median time to ANC recovery ≥ 0.5K/ul and platelet recovery ≥ 50 K/ul was 33 days (range 24-38) and 29 days (range 17-55), respectively; at VEN 400 (n=3) 29 days (range 22-34) and 24 days (23-25), respectively. All 10 evaluable pts responded (9CR/1CRi); each achieved CR with a single induction cycle. 6/8 assessed achieved MRD negativity by flow cytometry. The table lists the specific pt characteristics, adverse events and outcome. Conclusions: VEN 400 mg/d d1-11 with "3+7" induction starting on d 2 can be given safely in pts age 18-60 with newly diagnosed AML. The regimen appears highly active. Moving forward, pts will be treated with 3+7/VEN 400 induction followed by high dose cytarabine starting on day 2 plus VEN 200 (and 400 or 600 potential cohorts) d1-8. Prophylactic anti-gut flora antibiotics will accompany all courses. The 3+7/VEN 400 induction regimen derived from this trial will be compared to 3+7 in non-favorable risk newly diagnosed AML pts ages 18-60 and VEN/high dose cytarabine post-remission therapy evaluated in the context of North American Intergroup studies. Table Disclosures Stone: Stemline: Consultancy; Astellas: Consultancy; Novartis: Consultancy, Research Funding; Agios: Consultancy, Research Funding; Actinium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Other: DSMB; Syntrix: Other: DSMB; Syndax: Consultancy, Research Funding; Trovagene: Consultancy; Biolinerx: Consultancy; Macrogenics: Consultancy; Argenix: Other; Jazz: Consultancy; Janssen: Consultancy; Aztra-Zeneca: Consultancy; Celgene: Consultancy, Other; Pfizer: Consultancy; Daiichi-Sankyo: Consultancy; Arog: Consultancy, Research Funding; Gemoab: Consultancy; Syros: Consultancy; Abbvie: Consultancy, Research Funding. DeAngelo:Forty-Seven: Consultancy; Blueprint Medicines Corporation: Consultancy, Research Funding; Agios: Consultancy; Autolos: Consultancy; Amgen: Consultancy; Incyte Corporation: Consultancy; Glycomimetics: Research Funding; Abbvie: Research Funding; Takeda: Consultancy; Shire: Consultancy; Pfizer: Consultancy; Novartis: Consultancy, Research Funding; Jazz: Consultancy. Letai:Dialectic: Membership on an entity's Board of Directors or advisory committees; Flash Therapeutics: Membership on an entity's Board of Directors or advisory committees; Zentalis: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Consultancy; AbbVie: Consultancy; Novartis: Research Funding; Chugai: Other: Lecture Fees. Konopleva:Ablynx: Research Funding; Calithera: Research Funding; Rafael Pharmaceutical: Research Funding; Cellectis: Research Funding; AstraZeneca: Research Funding; Forty-Seven: Consultancy, Research Funding; Reata Pharmaceutical Inc.;: Patents & Royalties: patents and royalties with patent US 7,795,305 B2 on CDDO-compounds and combination therapies, licensed to Reata Pharmaceutical; Agios: Research Funding; Amgen: Consultancy; Kisoji: Consultancy; F. Hoffmann La-Roche: Consultancy, Research Funding; Stemline Therapeutics: Consultancy, Research Funding; Ascentage: Research Funding; Eli Lilly: Research Funding; Sanofi: Research Funding; AbbVie: Consultancy, Research Funding; Genentech: Consultancy, Research Funding. Garcia:Genentech: Research Funding; Eli Lily: Research Funding; Pfizer: Research Funding; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. OffLabel Disclosure: Venetoclax used with standard AML induction therapy
- Published
- 2020
43. Multiplexed CRISPR In Vivo Editing of CLL Loss-of-Function Lesions Models Transformation of Chronic Lymphocytic Leukemia into Richter's Syndrome
- Author
-
Lili Wang, Elisa Ten Hacken, Elizabeth Witten, Catherine J. Wu, Shuqiang Li, Kaitlyn Baranowski, Donna Neuberg, Geoffrey Fell, Robert A. Redd, Michaela Gruber, Luca Pinello, Leah Billington, Mohamed Uduman, Ruben D. Carrasco, Tomasz Sewastianik, Erin M. Parry, María Hernández-Sánchez, Shanye Yin, Kendell Clement, and Kenneth J. Livak
- Subjects
Lineage (genetic) ,medicine.diagnostic_test ,Chronic lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Flow cytometry ,Pathogenesis ,medicine.anatomical_structure ,Cancer research ,medicine ,Progenitor cell ,CD5 ,Gene ,B cell - Abstract
Although we have gained a wealth of knowledge from large-scale DNA sequencing studies across blood cancers, we still know little about the functional interplay of the discovered putative drivers in the generation of chronic lymphocytic leukemia (CLL) and its transformation into Richter's syndrome (RS). We have previously observed that CRISPR-Cas9 in vivo B-cell editing of common CLL loss-of-function (LOF) lesions (Atm, Trp53, Chd2, Birc3, Mga, Samhd1) can increase in vitro B cell fitness, but is not sufficient to sustain in vivo B cell survival after 12 months post-transplant. We therefore asked whether combinatorial introduction of mutations was required for CLL development. To this end, we generated transplant lines by in vitro engineering of early stem and progenitor cells (Lineage- Sca-1+ c-kit+ [LSK]) from MDR-/-Cd19-Cas9 donor mice (animals expressing Cas9-GFP in a B-cell restricted fashion and the leukemogenic homozygous MDR lesion, mimicking del(13q)) with pooled lentivirus expressing sgRNAs against the 6 genes of interest and the mCherry marker. Engineered LSKs were then re-transplanted into sub-lethally irradiated immune-competent CD45.1 or immune-deficient NSG recipients (n=35/strain). Parallel control cohorts of equal size were generated by transducing LSKs with a pool of 6 non-targeting sgRNAs. Disease development (B220+CD5+Igk+ cells) was assessed by flow cytometric analysis of bi-monthly peripheral bleeds, starting at 4 months post-transplant, and flow cytometry/IHC were utilized to classify tumors at euthanasia. Analysis of PCR-based targeted NGS of peripheral blood edited tumor cells (GFP+mCherry+) was performed utilizing CRISPResso software to assess presence of the 6 LOF mutations. We observed incidence of circulating CLL in 28/35 (80%) CD45.1 and 27/35 (77%) NSG mice, whereas only 5/35 (14%) CD45.1 and 4/35 (11.4%) NSG from the non-targeting control cohort developed CLL-like disease (P To determine the genetic composition of the 55 leukemias/lymphomas (n=22, 11 and 22, with patterns A, B and C), we interrogated the LOF mutational burden at euthanasia. We observed a median number of 4 LOF mutations (range:1-6), and a high overall frequency of Trp53 lesions (58%). The other 5 drivers were less prevalent (Mga: 26%; Chd2: 21%; Samhd1: 17%; Birc3 and Atm: 13%). Trp53 mutations were predominantly clonal (≥90% indels, P In conclusion, we demonstrate that combinatorial in vivo modeling of CLL-LOF mutations leads not only to CLL development, but also to RS, thus establishing a faithful framework for analysis of genetic and microenvironmental determinants of disease transformation. We are now interrogating genome-wide mutational patterns and clonal architecture of CLL vs. RS, while analyzing their microenvironmental composition. These novel models provide a unique platform to discern critical insights into RS pathogenesis and to discover RS-specific therapeutic vulnerabilities. Disclosures Clement: Edilytics: Current Employment, Current equity holder in private company. Wu:Pharmacyclics: Research Funding; BionTech: Current equity holder in publicly-traded company.
- Published
- 2020
44. Phase I Study of Ixazomib with Conventional Chemotherapy in the Treatment of Acute Myeloid Leukemia in Older Adults
- Author
-
Donna Neuberg, Rupa Narayan, Amir T. Fathi, Aura Y. Ramos, Julia Foster, Jenna A. Moran, Traci M. Blonquist, Meghan Bergeron, Philip C. Amrein, Eyal C. Attar, Megan K. Vartanian, Molly Macrae, Christina Bertoli, Geoffrey Fell, Steven L. McAfee, Gabriela S. Hobbs, Hanno Hock, Andrew M. Brunner, Tina T. Som, Meghan Burke, Kristin McGregor, Tanya T. Behnan, and Jennifer Lombardi Story
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Bortezomib ,Myelodysplastic syndromes ,Standard treatment ,Immunology ,Consolidation Chemotherapy ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Ixazomib ,chemistry.chemical_compound ,chemistry ,Hypomethylating agent ,Internal medicine ,Cohort ,medicine ,Cytarabine ,business ,medicine.drug - Abstract
Introduction: Outcomes for acute myeloid leukemia (AML) among older patients has remained largely unchanged for decades. Long-term survival for patients aged >60 years is poor (median survival 10.5 months). Targeting the proteasome in AML is attractive, since leukemia stem cells have demonstrated sensitivity to proteasome inhibition in preclinical models, perhaps through down regulation of nuclear NF-KB (Guzman, Blood 2001). AML cell lines are susceptible to synergistic cytotoxicity when bortezomib, a proteasome inhibitor, is combined with daunorubicin and cytarabine. We have shown that adding bortezomib to standard treatment in AML results in a high remission rate, although grade 2 sensory neurotoxicity was noted in approximately 12% of treated patients. A newer generation proteasome inhibitor, ixazomib, is less frequently associated with neurotoxicity, and, therefore, was selected for combination with conventional chemotherapy in this phase I trial. The primary objective of this study was to determine the maximum tolerated dose (MTD) of ixazomib in combination with conventional induction and consolidation chemotherapy for AML. Herein are the initial results of this trial. Methods: Adults >60 years of age with newly diagnosed AML were screened for eligibility. Patients with secondary AML were eligible, including those with prior hypomethylating agent therapy for myelodysplastic syndromes (MDS). We excluded those with promyelocytic leukemia. There were 2 phases in this study. In the first phase (A), the induction treatment consisted of the following: cytarabine 100 mg/m2/day by continuous IV infusion, Days 1-7; daunorubicin 60 mg/m2/day IV, Days 1, 2, 3, and ixazomib was provided orally at the cohort dose, Days 2, 5, 9, and 12. Consolidaton or transplant was at the discretion of the treating physician in phase A. In the second phase (B), induction was the same as that with the determined MTD of ixazomib. All patients were to be treated with the following consolidation: cytarabine at 2 g/m2/day, days 1-5 with ixazomib on days 2, 5, 9, and 12 at the cohort dose for consolidation. A standard 3 + 3 patient cohort dose escalation design was used to determine whether the dose of ixazomib could be safely escalated in 3 cohorts (1.5 mg/day, 2.3 mg/day, 3.0 mg/day), initially in induction (phase A) and subsequently in consolidation (phase B). The determined MTD of ixazomib in the first portion (A) of the trial was used during induction in the second portion (B), which sought to determine the MTD for ixazomib during consolidation. Secondary objectives included rate of complete remission, disease-free survival, and overall survival (OS). Results: Thirty-six patients have been enrolled on study, and 28 have completed dose levels A-1 through A-3 and B1 through B-2. Full information on cohort B-3 has not yet been obtained, hence, this report covers the experience with the initial 28 patients, cohorts A-1 through B-2. There were 12 (43%) patients among the 28 with secondary AML, either with prior hematologic malignancy or therapy-related AML. Nineteen patients (68%) were male, and the median age was 68 years (range 61-80 years). There have been no grade 5 toxicities due to study drug. Three patients died early due to leukemia, 2 of which were replaced for assessment of the MTD. Nearly all the grade 3 and 4 toxicities were hematologic (Table). There was 1 DLT (grade 4 platelet count decrease extending beyond Day 42). There has been no grade 3 or 4 neurotoxicity with ixazomib to date. Among the 28 patients in the first 5 cohorts, 22 achieved complete remissions (CR) and 2 achieved CRi, for a composite remission rate (CCR) of 86%. Among the 12 patients with secondary AML 8 achieved CR and 2 achieved CRi, for a CCR of 83%. The median OS for the 28 patients has not been reached (graph). The 18-month OS estimate was 65% [90% CI, 50-85%]. Conclusions: The highest dose level (3 mg) of ixazomib planned for induction in this trial has been reached safely. For consolidation there have been no serious safety issues in the first 2 cohorts with a dose up to 2.3 mg, apart from 1 DLT in the form of delayed platelet count recovery. The recommended phase 2 dose of ixazomib for induction is 3 mg. Accrual to cohort B-3 is ongoing. Notably, to date, no grade 3 or 4 neurotoxicity has been encountered. The remission rate in this older adult population with the addition of ixazomib to standard chemotherapy appears favorable. Figure Disclosures Amrein: Amgen: Research Funding; AstraZeneca: Consultancy, Research Funding; Takeda: Research Funding. Attar:Aprea Therapeutics: Current Employment. Brunner:Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Forty-Seven Inc: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Research Funding; Takeda: Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding. Hobbs:Constellation: Honoraria, Research Funding; Novartis: Honoraria; Incyte: Research Funding; Merck: Research Funding; Bayer: Research Funding; Jazz: Honoraria; Celgene/BMS: Honoraria. Neuberg:Celgene: Research Funding; Madrigak Pharmaceuticals: Current equity holder in publicly-traded company; Pharmacyclics: Research Funding. Fathi:Blueprint: Consultancy; Boston Biomedical: Consultancy; BMS/Celgene: Consultancy, Research Funding; Novartis: Consultancy; Kura Oncology: Consultancy; Trillium: Consultancy; Amgen: Consultancy; Seattle Genetics: Consultancy, Research Funding; Abbvie: Consultancy; Pfizer: Consultancy; Newlink Genetics: Consultancy; Forty Seven: Consultancy; Trovagene: Consultancy; Kite: Consultancy; Daiichi Sankyo: Consultancy; Astellas: Consultancy; Amphivena: Consultancy; PTC Therapeutics: Consultancy; Agios: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Jazz: Consultancy. OffLabel Disclosure: Ixazomib is FDA approved for multiple myeloma. We are using it in this trial for acute myeloid leukemia.
- Published
- 2020
45. An endogenous glucocorticoid-cytokine signaling circuit promotes CD8+ T cell dysfunction in the tumor microenvironment
- Author
-
Max Klapholz, Karen O. Dixon, Vijay K. Kuchroo, Ana C. Anderson, Elena Christian, Davide Mangani, Geoffrey Fell, Donna Neuberg, Junrong Xia, Katherine Tooley, Aviv Regev, Nandini Acharya, Orit Rozenblatt-Rosen, Asaf Madi, Meromit Singer, and Huiyuan Zhang
- Subjects
0303 health sciences ,Tumor microenvironment ,business.industry ,Melanoma ,medicine.medical_treatment ,medicine.disease ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Cytokine ,Glucocorticoid receptor ,030220 oncology & carcinogenesis ,Cancer research ,Cytotoxic T cell ,Medicine ,business ,Receptor ,Glucocorticoid ,CD8 ,030304 developmental biology ,medicine.drug - Abstract
Identifying signals in the tumor microenvironment (TME) that promote CD8+ T cell dysfunction can inform improved therapeutic approaches for cancer. Here, we identify that Nr3c1, the gene encoding the glucocorticoid receptor (GR), is highly expressed in dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). The GR transactivates expression of multiple checkpoint receptors and loss of GR in CD8+ T cells limits dysfunctional phenotype in CD8+ TILs resulting in improved tumor growth control. We show that glucocorticoids can be produced in the TME and that they co-operate with the immunosuppressive cytokine IL-27 to promote the dysfunction gene program in CD8+ T cells. The presence of the glucocorticoid + IL27 signature in CD8+ TILs correlates with failure to respond to checkpoint blockade in melanoma patients, highlighting the relevance of this immunoregulatory glucocorticoid-cytokine circuit in tumor tissue.
- Published
- 2019
- Full Text
- View/download PDF
46. Alisertib plus induction chemotherapy in previously untreated patients with high-risk, acute myeloid leukaemia: a single-arm, phase 2 trial
- Author
-
Gabriela S. Hobbs, Kristin McGregor, Donna Neuberg, R. Coleman Lindsley, Nicole Nelson, Daniel J. DeAngelo, Margaret C. Wey, Richard Stone, Kaitlyn M. Fishman, Philip C. Amrein, Jessica Rae, Jacqueline S. Garcia, Emma Logan, Traci M. Blonquist, Amity L. Manning, Myrna Nahas, Aura Y. Ramos, David P. Steensma, Lourdes M. Mendez, Meghan Bergeron, Tina T. Som, Meghan Burke, Jenna A. Moran, Yi Bin Chen, Julia Foster, Steven L. McAfee, Andrew M. Brunner, Molly Macrae, Hanno Hock, Eric S. Winer, Marlise R. Luskin, David Avigan, Karen K. Ballen, Michele Baltay, Jacalyn Rosenblatt, Nicole M. Hermance, Malgorzata McMasters, Timothy A. Graubert, Frank C. Kuo, Jennifer Lombardi Story, Amir T. Fathi, Geoffrey Fell, Christina Bertoli, Tanya T. Behnan, and Christine Connolly
- Subjects
Male ,medicine.medical_specialty ,Myeloid ,Neutropenia ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Risk Factors ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Idarubicin ,Humans ,Myeloproliferative neoplasm ,Aged ,business.industry ,Cytarabine ,Induction chemotherapy ,Hematology ,Azepines ,Induction Chemotherapy ,Middle Aged ,medicine.disease ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Pyrimidines ,chemistry ,030220 oncology & carcinogenesis ,Alisertib ,Female ,business ,Febrile neutropenia ,030215 immunology ,medicine.drug ,Follow-Up Studies - Abstract
Summary Background Increased aurora A kinase (AAK) expression occurs in acute myeloid leukaemia; AAK inhibition is a promising therapeutic target in this disease. We therefore aimed to assess the activity of alisertib combined with 7 + 3 induction chemotherapy in previously untreated patients with high-risk acute myeloid leukaemia. Methods We did a single-arm, phase 2 trial of patients recruited from the Dana–Farber/Harvard Cancer Center in the USA. Eligible patients had previously untreated acute myeloid leukaemia, an Eastern Cooperative Oncology Group performance status of 0–2, and were at high risk of disease as defined by the presence of an adverse-risk karyotype, the presence of secondary acute myeloid leukaemia arising from previous myelodysplastic syndrome or myeloproliferative neoplasm, the presence of therapy-related acute myeloid leukaemia, or being 65 years or older. Enrolled patients received 7 + 3 induction chemotherapy of continuous infusion of cytarabine (100 mg/m2 per day on days 1–7) and intravenous bolus of idarubicin (12 mg/m2 per day on days 1–3). Oral alisertib (30 mg) was given twice per day on days 8–15. Patients could receive up to four consolidation cycles with cytarabine and alisertib, and alisertib maintenance for 12 months. The primary endpoint was a composite including the proportion of patients achieving complete remission and those with a complete remission with incomplete neutrophil or platelet count recovery. Analyses were per-protocol. This study is registered with Clinicaltrials.gov , number NCT02560025 , and has completed enrolment. Findings Between Dec 31, 2015, and Aug 1, 2017, we enrolled a total of 39 eligible patients. 19 (49%) of 39 patients had secondary acute myeloid leukaemia and three (8%) had therapy-related acute myeloid leukaemia. At mid-induction, 33 (85%) of 39 patients showed marrow aplasia, six (15%) received re-induction. The median follow-up was 13·7 months (IQR 12·7–14·4). Composite remission was 64% (two-stage 95% CI 48–79), with 20 (51%) of 39 patients achieving complete remission and five (13%) achieving complete remission with incomplete neutrophil or platelet count recovery. The most common grade 3 or 4 adverse events included febrile neutropenia (16 [41%] of 39), neutropenia (12 [31%]), thrombocytopenia (13 [33%]), anaemia (11 [28%]), anorexia (nine [23%]), and oral mucositis (four [10%]). No treatment-related deaths were observed. Interpretation These results suggest that alisertib combined with induction chemotherapy is active and safe in previously untreated patients with high-risk acute myeloid leukaemia. This study met criteria to move forward to a future randomised trial. Funding Millennium Pharmaceuticals.
- Published
- 2019
47. To randomize, or not to randomize, that is the question: using data from prior clinical trials to guide future designs
- Author
-
Brian M. Alexander, Lorenzo Trippa, Patrick Y. Wen, Geoffrey Fell, Alyssa M Vanderbeek, Rifaquat Rahman, Timothy F. Cloughesy, and Steffen Ventz
- Subjects
Cancer Research ,Standard of care ,Randomization ,Computer science ,Clinical study design ,Outcome (probability) ,law.invention ,Clinical trial ,Metadata Analysis ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Randomized controlled trial ,law ,Research Design ,030220 oncology & carcinogenesis ,Statistics ,Clinical endpoint ,Benchmark (computing) ,Humans ,030212 general & internal medicine ,Neurology (clinical) ,Randomized Controlled Trials as Topic - Abstract
Background Understanding the value of randomization is critical in designing clinical trials. Here, we introduce a simple and interpretable quantitative method to compare randomized designs versus single-arm designs using indication-specific parameters derived from the literature. We demonstrate the approach through application to phase II trials in newly diagnosed glioblastoma (ndGBM). Methods We abstracted data from prior ndGBM trials and derived relevant parameters to compare phase II randomized controlled trials (RCTs) and single-arm designs within a quantitative framework. Parameters included in our model were (i) the variability of the primary endpoint distributions across studies, (ii) potential for incorrectly specifying the single-arm trial’s benchmark, and (iii) the hypothesized effect size. Strengths and weaknesses of RCT and single-arm designs were quantified by various metrics, including power and false positive error rates. Results We applied our method to show that RCTs should be preferred to single-arm trials for evaluating overall survival in ndGBM patients based on parameters estimated from prior trials. More generally, for a given effect size, the utility of randomization compared with single-arm designs is highly dependent on (i) interstudy variability of the outcome distributions and (ii) potential errors in selecting standard of care efficacy estimates for single-arm studies. Conclusions A quantitative framework using historical data is useful in understanding the utility of randomization in designing prospective trials. For typical phase II ndGBM trials using overall survival as the primary endpoint, randomization should be preferred over single-arm designs.
- Published
- 2019
48. Progression Risk Stratification of Asymptomatic Waldenström Macroglobulinemia
- Author
-
David Liu, Kalvis Hornburg, Romanos Sklavenitis-Pistofidis, Irene M. Ghobrial, Stephen M. Ansell, Jorge J. Castillo, Maria Gavriatopoulou, Saurabh Zanwar, Efstathios Kastritis, Geoffrey Fell, Joseph Cappuccio, Lorenzo Trippa, Robert A. Kyle, Robert J. Soiffer, Carl Jannes Neuse, Henry Dumke, Jithma P. Abeykoon, Mark Bustoros, Catherine R. Marinac, Jenny Soiffer, Meletios A. Dimopoulos, Chia Jen Liu, Steven P. Treon, Prashant Kapoor, Cody J. Boehner, and Kaitlen Reyes
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Time Factors ,Serum Albumin, Human ,Disease ,Gastroenterology ,Asymptomatic ,Risk Assessment ,Decision Support Techniques ,Bone Marrow ,Predictive Value of Tests ,Risk Factors ,Internal medicine ,Medicine ,Humans ,Asymptomatic Diseases ,Aged ,Aged, 80 and over ,business.industry ,Macroglobulinemia ,Waldenstrom macroglobulinemia ,ORIGINAL REPORTS ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Oncology ,Immunoglobulin M ,Predictive value of tests ,Risk stratification ,Mutation ,Myeloid Differentiation Factor 88 ,Disease Progression ,Female ,medicine.symptom ,Waldenstrom Macroglobulinemia ,Risk assessment ,business ,beta 2-Microglobulin ,Biomarkers ,Boston - Abstract
BACKGROUND Waldenström macroglobulinemia (WM) is preceded by asymptomatic WM (AWM), for which the risk of progression to overt disease is not well defined. METHODS We studied 439 patients with AWM, who were diagnosed and observed at Dana-Farber Cancer Institute between 1992 and 2014. RESULTS During the 23-year study period, with a median follow-up of 7.8 years, 317 patients progressed to symptomatic WM (72%). Immunoglobulin M 4,500 mg/dL or greater, bone marrow lymphoplasmacytic infiltration 70% or greater, β2-microglobulin 4.0 mg/dL or greater, and albumin 3.5 g/dL or less were all identified as independent predictors of disease progression. To assess progression risk in patients with AWM, we trained and cross-validated a proportional hazards model using bone marrow infiltration, immunoglobulin M, albumin, and beta-2 microglobulin values as continuous measures. The model divided the cohort into three distinct risk groups: a high-risk group with a median time to progression (TTP) of 1.8 years, an intermediate-risk group with a median TTP of 4.8 years, and a low-risk group with a median TTP of 9.3 years. We validated this model in two external cohorts, demonstrating robustness and generalizability. For clinical applicability, we made the model available as a Web page application ( www.awmrisk.com ). By combining two cohorts, we were powered to identify wild type MYD88 as an independent predictor of progression (hazard ratio, 2.7). CONCLUSION This classification system is positioned to inform patient monitoring and care and, for the first time to our knowledge, to identify patients with high-risk AWM who may need closer follow-up or benefit from early intervention.
- Published
- 2019
49. Prediction of Outcomes with a Computational Biology Model in Newly Diagnosed Glioblastoma Patients Treated with Radiation Therapy and Temozolomide
- Author
-
Brian M. Alexander, Neeraj Kumar Singh, Yatin Mundkur, Geoffrey Fell, Zakir Husain, Shireen Vali, Lorenzo Trippa, Keith L. Ligon, Taher Abbasi, Rifaquat Rahman, Anay Talawdekar, Patrick Y. Wen, and S. Alden
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_treatment ,MEDLINE ,Gene Dosage ,Computational biology ,Models, Biological ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,Temozolomide ,Medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Precision Medicine ,Antineoplastic Agents, Alkylating ,Aged ,Proportional Hazards Models ,Retrospective Studies ,Aged, 80 and over ,Radiation ,Proportional hazards model ,business.industry ,Brain Neoplasms ,Computational Biology ,Retrospective cohort study ,Middle Aged ,Precision medicine ,Combined Modality Therapy ,Survival Analysis ,Progression-Free Survival ,Clinical trial ,Radiation therapy ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Female ,Personalized medicine ,business ,Glioblastoma ,medicine.drug - Abstract
Precision medicine has been most successful in targeting single mutations, but personalized medicine using broader genomic tumor profiles for individual patients is less well developed. We evaluate a genomics-informed computational biology model (CBM) to predict outcomes from standard treatments and to suggest novel therapy recommendations in glioblastoma (GBM).In this retrospective study, 98 patients with newly diagnosed GBM undergoing surgery followed by radiation therapy and temozolomide at a single institution with available genomic data were identified. Incorporating mutational and copy number aberration data, a CBM was used to simulate the response of GBM tumor cells and generate efficacy predictions for radiation therapy (RTeff) and temozolomide (TMZeff). RTeff and TMZeff were evaluated for association with overall survival and progression-free survival in a Cox regression model. To demonstrate a CBM-based individualized therapy strategy, treatment recommendations were generated for each patient by testing a panel of 45 central nervous system-penetrant US Food and Drug Administration-approved agents.High RTeff scores were associated with longer survival on univariable analysis (P.001), which persisted after controlling for age, extent of resection, performance status, MGMT, and IDH status (P = .017). High RTeff patients had a longer overall survival compared with low RTeff patients (median, 27.7 vs 14.6 months). High TMZeff was also associated with longer survival on univariable analysis (P = .007) but did not hold on multivariable analysis, suggesting an interplay with MGMT status. Among predictions of the 3 most efficacious combination therapies for each patient, only 2.4% (7 of 294) of 2-drug recommendations produced by the CBM included TMZ.CBM-based predictions of RT and TMZ effectiveness were associated with survival in patients with newly diagnosed GBM treated with those therapies, suggesting a possible predictive utility. Furthermore, the model was able to suggest novel individualized monotherapies and combinations. Prospective evaluation of such a personalized treatment strategy in clinical trials is needed.
- Published
- 2019
50. Deviation from the Proportional Hazards Assumption in Randomized Phase 3 Clinical Trials in Oncology: Prevalence, Associated Factors, and Implications
- Author
-
Alyssa M. Vanderbeek, Geoffrey Fell, Lorenzo Trippa, Brian M. Alexander, Andrea Arfè, Rifaquat Rahman, and Steffen Ventz
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,MEDLINE ,Kaplan-Meier Estimate ,Analysis of clinical trials ,01 natural sciences ,010104 statistics & probability ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Neoplasms ,medicine ,Humans ,0101 mathematics ,Precision Medicine ,Proportional Hazards Models ,Randomized Controlled Trials as Topic ,business.industry ,Proportional hazards model ,Cancer ,Odds ratio ,Precision medicine ,medicine.disease ,Confidence interval ,Clinical trial ,Clinical Trials, Phase III as Topic ,030220 oncology & carcinogenesis ,Immunotherapy ,business - Abstract
Purpose: Deviations from proportional hazards (DPHs), which may be more prevalent in the era of precision medicine and immunotherapy, can lead to underpowered trials or misleading conclusions. We used a meta-analytic approach to estimate DPHs across cancer trials, investigate associated factors, and evaluate data-analysis approaches for future trials. Experimental Design: We searched PubMed for phase III trials in breast, lung, prostate, and colorectal cancer published in a preselected list of journals between 2014 and 2016 and extracted individual patient-level data (IPLD) from Kaplan–Meier curves. We re-analyzed IPLD to identify DPHs. Potential efficiency gains, when DPHs were present, of alternative statistical methods relative to standard log-rank based analysis were expressed as sample-size requirements for a fixed power level. Results: From 152 trials, we obtained IPLD on 129,401 patients. Among 304 Kaplan–Meier figures, 75 (24.7%) exhibited evidence of DPHs, including eight of 14 (57%) KM pairs from immunotherapy trials. Trial type [immunotherapy, odds ratio (OR), 4.29; 95% confidence interval (CI), 1.11–16.6], metastatic patient population (OR, 3.18; 95% CI, 1.26–8.05), and non-OS endpoints (OR, 3.23; 95% CI, 1.79–5.88) were associated with DPHs. In immunotherapy trials, alternative statistical approaches allowed for more efficient clinical trials with fewer patients (up to 74% reduction) relative to log-rank testing. Conclusions: DPHs were found in a notable proportion of time-to-event outcomes in published clinical trials in oncology and was more common for immunotherapy trials and non-OS endpoints. Alternative statistical methods, without proportional hazards assumptions, should be considered in the design and analysis of clinical trials when the likelihood of DPHs is high.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.