69 results on '"Patricia Kropf"'
Search Results
2. Supplementary Table S1 from Safety, Outcomes, and T-Cell Characteristics in Patients with Relapsed or Refractory MDS or CMML Treated with Atezolizumab in Combination with Guadecitabine
- Author
-
Kirsten Grønbaek, Imran Siddiqi, Stephen B. Baylin, Peter Jones, Jean-Pierre Issa, Sine Reker Hadrup, Benjamin A. Youngblood, Galen Hostetter, Ryan Burgos, Kristin Horwood, Brett T. Brinker, Jack Cowland, Staffan Holmberg-Thyden, Ashkan Emadi, Hyo Sik Jang, Denice Tsao-Wei, Jakob Werner Hansen, Patricia Kropf, Vu H. Duong, Sunil Kumar Saini, Andreas Due Ørskov, Maria R. Baer, and Casey L. O'Connell
- Abstract
Specific genes sequenced in bone marrow mononuclear cells.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Methods from Safety, Outcomes, and T-Cell Characteristics in Patients with Relapsed or Refractory MDS or CMML Treated with Atezolizumab in Combination with Guadecitabine
- Author
-
Kirsten Grønbaek, Imran Siddiqi, Stephen B. Baylin, Peter Jones, Jean-Pierre Issa, Sine Reker Hadrup, Benjamin A. Youngblood, Galen Hostetter, Ryan Burgos, Kristin Horwood, Brett T. Brinker, Jack Cowland, Staffan Holmberg-Thyden, Ashkan Emadi, Hyo Sik Jang, Denice Tsao-Wei, Jakob Werner Hansen, Patricia Kropf, Vu H. Duong, Sunil Kumar Saini, Andreas Due Ørskov, Maria R. Baer, and Casey L. O'Connell
- Abstract
Isolation and DNA extraction of bone marrow cell populations; Targeted DNA sequencing; Peripheral blood mononuclear cell (PBMC) staining and flow cytometry analysis
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Safety, Outcomes, and T-Cell Characteristics in Patients with Relapsed or Refractory MDS or CMML Treated with Atezolizumab in Combination with Guadecitabine
- Author
-
Kirsten Grønbaek, Imran Siddiqi, Stephen B. Baylin, Peter Jones, Jean-Pierre Issa, Sine Reker Hadrup, Benjamin A. Youngblood, Galen Hostetter, Ryan Burgos, Kristin Horwood, Brett T. Brinker, Jack Cowland, Staffan Holmberg-Thyden, Ashkan Emadi, Hyo Sik Jang, Denice Tsao-Wei, Jakob Werner Hansen, Patricia Kropf, Vu H. Duong, Sunil Kumar Saini, Andreas Due Ørskov, Maria R. Baer, and Casey L. O'Connell
- Abstract
Purpose:We hypothesized that resistance to hypomethylating agents (HMA) among patients with myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) would be overcome by combining a programmed death-ligand 1 antibody with an HMA.Patients and Methods:We conducted a Phase I/II, multicenter clinical trial for patients with MDS not achieving an International Working Group response after at least 4 cycles of an HMA (“refractory”) or progressing after a response (“relapsed”) with 3+ or higher risk MDS by the revised International Prognostic Scoring System (IPSS-R) and CMML-1 or -2. Phase I consisted of a 3+3 dose-escalation design beginning with guadecitabine at 30 mg/m2 and escalating to 60 mg/m2 Days 1 to 5 with fixed-dose atezolizumab: 840 mg intravenously Days 8 and 22 of a 28-day cycle. Primary endpoints were safety and tolerability; secondary endpoints were overall response rate (ORR) and survival.Results:Thirty-three patients, median age 73 (range 54–85), were treated. Thirty patients had MDS and 3 had CMML, with 30% relapsed and 70% refractory. No dose-limiting toxicities were observed in Phase I. There were 3 (9%) deaths in ≤ 30 days. Five patients (16%) came off study for drug-related toxicity. Immune-related adverse events (IRAE) occurred in 12 (36%) patients (4 grade 3, 3 grade 2, and 5 grade1). ORR was 33% [95% confidence interval (CI), 19%–52%] with 2 complete remission (CR), 3 hematologic improvement, 5 marrow CR, and 1 partial remission. Median overall survival was 15.1 (95% CI, 8.5–25.3) months.Conclusions:Guadecitabine with atezolizumab has modest efficacy with manageable IRAEs and typical cytopenia-related safety concerns for patients with relapsed or refractory MDS and CMML.
- Published
- 2023
- Full Text
- View/download PDF
5. The Effect of Mobilizing Large Numbers of Cd34+ Cells (Super-Mobilizers) on the Engraftment and Survival in Patients Undergoing Autologous Stem Cell Transplantation
- Author
-
Jan Moreb, Lori Lantos, Franklin Chen, Kathleen Elliott, James Dugan, Alan Skarbnik, Patricia Kropf, and Kimberly Ward
- Published
- 2023
- Full Text
- View/download PDF
6. Guadecitabine (SGI-110) in patients with intermediate or high-risk myelodysplastic syndromes: phase 2 results from a multicentre, open-label, randomised, phase 1/2 trial
- Author
-
Mohammad Azab, Scott D. Lunin, Patricia Kropf, Nikolai A. Podoltsev, Jesus G. Berdeja, Michael R. Savona, Todd L. Rosenblat, Harold N. Keer, Hagop M. Kantarjian, Katherine Walsh, Guillermo Garcia-Manero, Wendy Stock, Elias Jabbour, Raoul Tibes, Jean Pierre J. Issa, Karen Yee, Joseph R. Mace, Ellen K. Ritchie, Elizabeth A. Griffiths, Casey O'Connell, Gail J. Roboz, and Yong Hao
- Subjects
Adult ,Male ,medicine.medical_specialty ,Neutropenia ,Adolescent ,Decitabine ,Antineoplastic Agents ,Severity of Illness Index ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,Adverse effect ,Survival rate ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,business.industry ,Myelodysplastic syndromes ,Hematology ,Middle Aged ,medicine.disease ,Thrombocytopenia ,Survival Rate ,Treatment Outcome ,Tolerability ,Hypomethylating agent ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Azacitidine ,Female ,business ,030215 immunology ,medicine.drug - Abstract
Summary Background Guadecitabine is a next-generation hypomethylating agent whose active metabolite decitabine has a longer in-vivo exposure time than intravenous decitabine. More effective hypomethylating agents are needed for the treatment of myelodysplastic syndromes. In the present study, we aimed to compare the activity and safety of two doses of guadecitabine in hypomethylating agent treatment-naive or relapsed or refractory patients with intermediate-risk or high-risk myelodysplastic syndromes. Methods This phase 2 part of the phase 1/2, randomised, open-label study enrolled patients aged 18 years or older from 14 North American medical centres with International Prognostic Scoring System intermediate-1-risk, intermediate-2-risk, or high-risk myelodysplastic syndromes, or chronic myelomonocytic leukaemia. They were either hypomethylating agent treatment-naive or had relapsed or refractory disease after previous hypomethylating agent treatment as determined by the investigators' judgment. Eligible patients had Eastern Cooperative Oncology Group performance status of 0–2. Patients were randomly assigned (1:1) using a computer algorithm for dynamic randomisation to subcutaneous guadecitabine 60 or 90 mg/m2 on days 1–5 of a 28-day treatment cycle. Treatment was stratified by previous treatment with hypomethylating agents and neither patients nor investigators were masked. The primary endpoint was overall response (a composite of complete response, partial response, marrow complete response, and haematological improvement) assessed in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01261312. Findings Between July 9, 2012, and April 7, 2014, 105 patients were enrolled: 55 (52%) were allocated to guadecitabine 60 mg/m2 (28 patients were treatment-naive and 27 had relapsed or refractory disease after previous hypomethylating agent treatment) and 50 (48%) patients to 90 mg/m2 (23 patients were treatment-naive and 27 had relapsed or refractory disease). Three (3%) patients of 105 did not receive study treatment and were excluded from analyses. Median follow-up was 3·2 years (IQR 2·8–3·5). The proportion of patients achieving an overall response did not significantly differ between dose groups (21 of 53 [40%, 95% CI 27–54] with 60 mg/m2 and 27 of 49 [55%, 95% CI 40–69] with 90 mg/m2; p=0·16). 25 of 49 (51%, 95% CI 36–66) patients who were treatment-naive and 23 of 53 (43%, 30–58) patients with relapsed or refractory disease achieved an overall response. The most common grade 3 or worse adverse events in both groups, regardless of relationship to treatment, were thrombocytopenia (22 [41%] of 53 patients in the 60 mg/m2 group and 28 [57%] of 49 in the 90 mg/m2 group), neutropaenia (21 [40%] and 25 [51%]), anaemia (25 [47%] and 24 [49%]), febrile neutropaenia (17 [32%] and 21 [43%]), and pneumonia (13 [25%] and 15 [31%]). Seven (7%) of 102 patients died due to adverse events (three with 90 mg/m2 and four with 60 mg/m2), and all except one were in the relapsed or refractory cohort. Two deaths were deemed treatment related (septic shock with 60 mg/m2; pneumonia with 90 mg/m2). Interpretation Guadecitabine was clinically active with acceptable tolerability in patients with intermediate-risk and high-risk myelodysplastic syndromes. Responses and overall survival in the relapsed or refractory cohort offer the potential of a new therapeutic option for patients for whom currently available hypomethylating agents are not successful. We therefore recommend guadecitabine at a dose of 60 mg/m2 on a 5-day schedule for these patients. Funding Astex Pharmaceuticals and Stand Up To Cancer.
- Published
- 2019
- Full Text
- View/download PDF
7. Functional genomic landscape of acute myeloid leukaemia
- Author
-
Robert H. Collins, Deirdre Devine, Beth Wilmot, Justin Ramsdill, Erik Segerdell, Bruno C. Medeiros, Brian J. Druker, Dylan Nelson, Micaela E. Martinez, Ryan C. Johnson, Robert Schuff, Robert P. Searles, Scott Weir, James Dibb, Elie Traer, Pierrette Lo, Haijiao Zhang, Rachel Henson, Tara A. Macey, Isabel English, Cody Coblentz, Christopher A. Eide, Ceilidh Nichols, Aurora Blucher, Ryan M. Winters, David L. Wiest, Corinne Visser, Michael W. Deininger, Stephen E. Kurtz, Daniel A. Pollyea, Justin M. Watts, Amy S. Carlos, Denise C. Connolly, Andy Kaempf, Angela Rofelty, Samuel B. Luty, Rachel J. Cook, Jill Peters, Kristen Werth, Shannon K. McWeeney, Joseph Carroll, Samantha L. Savage, Ronan T. Swords, Uma Borate, Aashis Thapa, Abdusebur Jemal, Joelle Wolf, Patricia Kropf, Rebecca Smith, Tyler Sweeney, Russell T. Burke, Rachel R. Pallapati, Anna Reister Schultz, Kim Hien T. Dao, Daniel Bottomly, Cristina E. Tognon, Alexey V. Danilov, Jason M. Glover, Jason D. MacManiman, Michie Degnin, Amy Yates, Libbey White, David K. Edwards, Anupriya Agarwal, Christopher R. Cogle, Kevin Watanabe-Smith, Leylah Drusbosky, Nicola Long, Motomi Mori, Christopher S. Hourigan, Tara L. Lin, Chenwei Lin, Jacqueline Martinez, Bill H. Chang, Richie Carpenter, Stephen E. Spurgeon, Brian Junio, Marc M. Loriaux, Craig T. Jordan, Hibery Ho, Selina Qiuying Liu, Melissa L. Abel, Amanda d’Almeida, Jake Wagner, Jade Bryant, Jeffrey W. Tyner, Jessica Leonard, and Kara Johnson
- Subjects
Male ,0301 basic medicine ,Myeloid ,Gene regulatory network ,Datasets as Topic ,Genomics ,Computational biology ,Biology ,Article ,DNA Methyltransferase 3A ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,medicine ,Humans ,Exome ,DNA (Cytosine-5-)-Methyltransferases ,Molecular Targeted Therapy ,Exome sequencing ,Regulation of gene expression ,Multidisciplinary ,Serine-Arginine Splicing Factors ,Genome, Human ,Sequence Analysis, RNA ,Nuclear Proteins ,medicine.disease ,Human genetics ,3. Good health ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Core Binding Factor Alpha 2 Subunit ,Female ,Nucleophosmin - Abstract
The implementation of targeted therapies for acute myeloid leukaemia (AML) has been challenging because of the complex mutational patterns within and across patients as well as a dearth of pharmacologic agents for most mutational events. Here we report initial findings from the Beat AML programme on a cohort of 672 tumour specimens collected from 562 patients. We assessed these specimens using whole-exome sequencing, RNA sequencing and analyses of ex vivo drug sensitivity. Our data reveal mutational events that have not previously been detected in AML. We show that the response to drugs is associated with mutational status, including instances of drug sensitivity that are specific to combinatorial mutational events. Integration with RNA sequencing also revealed gene expression signatures, which predict a role for specific gene networks in the drug response. Collectively, we have generated a dataset—accessible through the Beat AML data viewer (Vizome)—that can be leveraged to address clinical, genomic, transcriptomic and functional analyses of the biology of AML. Analyses of samples from patients with acute myeloid leukaemia reveal that drug response is associated with mutational status and gene expression; the generated dataset provides a basis for future clinical and functional studies of this disease.
- Published
- 2018
- Full Text
- View/download PDF
8. A phase-1 study of dasatinib plus all-trans retinoic acid in acute myeloid leukemia
- Author
-
Susan M. Christner, Seah H. Lim, Rafic Farah, Kathleen A. Dorritie, Michael Boyiadzis, Daniel P. Normolle, Alison R. Sehgal, Daniel Johnson, Annie Im, Jing-Zhao Hou, Patricia Kropf, Jan H. Beumer, Mounzer Agha, Anastasios Raptis, and Robert L. Redner
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Combination therapy ,Dasatinib ,Retinoic acid ,Tretinoin ,Drug Administration Schedule ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Adverse effect ,neoplasms ,Aged ,Aged, 80 and over ,business.industry ,Headache ,Myeloid leukemia ,Hematology ,Middle Aged ,Long QT Syndrome ,Retinoic acid receptor ,Treatment Outcome ,src-Family Kinases ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Leukemia, Myeloid ,030220 oncology & carcinogenesis ,Acute Disease ,Toxicity ,Cancer research ,business ,medicine.drug - Abstract
Src family kinases (SFKs) are hyperactivated in acute myeloid leukemia (AML). SFKs impede the retinoic acid receptor, and SFK inhibitors enhance all-trans retinoic acid (ATRA)-mediated cellular differentiation in AML cell lines and primary blasts. To translate these findings into the clinic, we undertook a phase I dose-escalation study of the combination of the SFK inhibitor dasatinib and ATRA in patients with high-risk myeloid neoplasms. Nine subjects were enrolled: six received 70 mg dasatinib plus 45 mg/m(2) ATRA daily, and three received 100 mg dasatinib plus 45 mg/m(2) ATRA daily for 28 days. Headache and QTc prolongations were the only two grade 3 adverse events observed. No significant clinical responses were observed. We conclude that the combination of 70 mg dasatinib and 45 mg/m(2) ATRA daily is safe with acceptable toxicity. Our results provide the safety profile for further investigations into the clinical efficacy of this combination therapy in myeloid malignancies.
- Published
- 2018
- Full Text
- View/download PDF
9. Demethylator phenotypes in acute myeloid leukemia
- Author
-
Charly R. Good, Andrew Kelly, Jozef Madzo, Jean Pierre J. Issa, Patricia Kropf, Priyanka Madireddi, and Jaroslav Jelinek
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,NPM1 ,Biology ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,AML ,medicine ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,DMP ,Epigenetics ,Aged ,Aged, 80 and over ,DNA methylation ,CIMP ,Cytogenetics ,Nuclear Proteins ,demethylation ,Myeloid leukemia ,Hematology ,Middle Aged ,Prognosis ,Phenotype ,Up-Regulation ,3. Good health ,Leukemia, Myeloid, Acute ,030104 developmental biology ,fms-Like Tyrosine Kinase 3 ,Oncology ,CpG site ,Mutation ,Fms-Like Tyrosine Kinase 3 ,Cancer research ,DNMT3A ,CpG Islands ,Female ,Nucleophosmin ,hypomethylation - Abstract
Acute myeloid leukemia (AML) often harbors mutations in epigenetic regulators, and also has frequent DNA hypermethylation, including the presence of CpG island methylator phenotypes (CIMPs). Although global hypomethylation is well known in cancer, the question of whether distinct demethylator phenotypes (DMPs) exist remains unanswered. Using Illumina 450k arrays for 194 patients from The Cancer Genome Atlas, we identified two distinct DMPs by hierarchical clustering: DMP.1 and DMP.2. DMP.1 cases harbored mutations in NPM1 (94%), FLT3 (71%) and DNMT3A (61%). Surprisingly, only 40% of patients with DNMT3A mutations were DMP.1, which has implications for mechanisms of transformation by this mutation. In contrast, DMP.2 AML was comprised of patients with t(8;21), inv(16) or t(15;17), suggesting common methylation defects connect these disparate rearrangements. RNA-seq revealed upregulated genes functioning in immune response (DMP.1) and development (DMP.2). We confirmed these findings by integrating independent 450k data sets (236 additional cases), and found prognostic effects by DMP status, independent of age and cytogenetics. The existence of DMPs has implications for AML pathogenesis and may augment existing tools in risk stratification.
- Published
- 2018
- Full Text
- View/download PDF
10. Dose, schedule, safety, and efficacy of guadecitabine in relapsed or refractory acute myeloid leukemia
- Author
-
Katherine Walsh, Casey O'Connell, David A. Rizzieri, Gail J. Roboz, Jean Pierre J. Issa, Hagop M. Kantarjian, James N. Lowder, Karen W.L. Yee, Yong Hao, Elias Jabbour, Woonbok Chung, Scott D. Lunin, Elizabeth A. Griffiths, Wendy Stock, Tania Curio, Ellen K. Ritchie, Patricia Kropf, Naval Daver, and Mohammad Azab
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Population ,Myeloid leukemia ,Cancer ,medicine.disease ,Transplantation ,03 medical and health sciences ,Regimen ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Hypomethylating agent ,030220 oncology & carcinogenesis ,Internal medicine ,Cohort ,medicine ,Adverse effect ,business ,education - Abstract
Background Outcomes for patients with relapsed or refractory acute myeloid leukemia (AML) are poor. Guadecitabine, a next-generation hypomethylating agent, could be useful in treating such patients. Methods In this multicenter, open-label, phase 2 dose-expansion study, AML patients from 10 North American medical centers were first randomized (1:1) to receive subcutaneous guadecitabine at 60 or 90 mg/m2 on 5 consecutive days in each 28-day cycle (5-day regimen). Subsequently, another cohort was treated for 10 days with 60 mg/m2 (10-day regimen). Results Between June 15, 2012, and August 19, 2013, 108 patients with previously treated AML consented to enroll in the study, and 103 of these patients were treated; 5 patients did not receive the study treatment. A total of 103 patients were included in the safety and efficacy analyses (24 and 26 patients who were randomly assigned to 60 and 90 mg/m2 /d, respectively [5-day regimen] and 53 patients who were assigned to 60 mg/m2 /d [10-day regimen]). The 90 mg/m2 dose showed no benefit in clinical outcomes in comparison with 60 mg/m2 in the randomized cohort. Composite complete response (CRc) and complete response (CR) rates were higher with the 10-day regimen versus the 5-day regimen (CRc, 30.2% vs 16.0%; P = .1061; CR, 18.9% vs 8%; P = .15). Adverse events (grade ≥ 3) were mainly hematologic, with a higher incidence on the 10-day regimen. Early all-cause mortality was low and similar between regimens. Twenty patients (8 on the 5-day regimen and 12 on the 10-day regimen) were bridged to hematopoietic cell transplantation. Conclusions Guadecitabine has promising clinical activity and an acceptable safety profile and thus warrants further development in this population. Cancer 2018;124:325-34. © 2017 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
- Published
- 2017
- Full Text
- View/download PDF
11. Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial
- Author
-
Karen W.L. Yee, Wendy Stock, Mohammad Azab, Katherine Walsh, Patricia Kropf, Michael R. Savona, Raoul Tibes, Todd L. Rosenblat, Elias Jabbour, Edwin P. Rock, Nikolai A. Podoltsev, Jesus G. Berdeja, Jean Pierre J. Issa, Farhad Ravandi, Hagop M. Kantarjian, Guillermo Garcia-Manero, David A. Rizzieri, Yong Hao, Casey O'Connell, Gail J. Roboz, and Elizabeth A. Griffiths
- Subjects
Adult ,0301 basic medicine ,medicine.medical_specialty ,Maximum Tolerated Dose ,Population ,Phases of clinical research ,Decitabine ,Kaplan-Meier Estimate ,Neutropenia ,Risk Assessment ,Disease-Free Survival ,Drug Administration Schedule ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Neoplasm Invasiveness ,Prospective Studies ,Infusions, Intravenous ,education ,Prospective cohort study ,Aged ,Neoplasm Staging ,Aged, 80 and over ,education.field_of_study ,Dose-Response Relationship, Drug ,business.industry ,Myelodysplastic syndromes ,Remission Induction ,Age Factors ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,Surgery ,Leukemia, Myeloid, Acute ,Regimen ,Treatment Outcome ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Azacitidine ,Patient Safety ,business ,Febrile neutropenia ,medicine.drug - Abstract
Summary Background The hypomethylating drugs azacitidine and decitabine have shown efficacy in myelodysplastic syndromes and acute myeloid leukaemia, but complete tumour responses are infrequent and of short duration, possibly because of the short half-lives and suboptimal bone marrow exposure of the drugs. Guadecitabine, a next-generation hypomethylating drug, has a longer half-life and exposure than its active metabolite decitabine. A phase 1 study established 60 mg/m 2 guadecitabine for 5 days as an effective treatment schedule. In this phase 2 study, we aimed to assess the safety and activity of two doses and schedules of guadecitabine in older (≥65 years) patients with treatment-naive acute myeloid leukaemia who were not candidates for intensive chemotherapy. Methods We did a multicentre, randomised, open-label, phase 1/2 study of guadecitabine in cohorts of patients with treatment-naive acute myeloid leukaemia, relapsed or refractory acute myeloid leukaemia, and myelodysplastic syndromes; here we report the phase 2 results from the cohort of treatment-naive patients with acute myeloid leukaemia. We included patients aged at least 65 years from 14 US medical centres (hospitals and specialist cancer clinics) who were not candidates for intensive chemotherapy and randomly assigned them (1:1) using a computer algorithm (for dynamic randomisation) to guadecitabine 60 or 90 mg/m 2 on days 1–5 (5-day schedule) of a 28-day treatment cycle. Treatment allocation was not masked. We also assigned additional patients to guadecitabine 60 mg/m 2 in a 10-day schedule in a 28-day treatment cycle after a protocol amendment. The primary endpoint was composite complete response (complete response, complete response with incomplete platelet recovery, or complete response with incomplete neutrophil recovery regardless of platelets). Response was assessed in all patients (as-treated) who received at least one dose of guadecitabine. We present the final analysis, although at the time of the database lock, 15 patients were still in follow-up for overall survival. This study is registered with ClinicalTrials.gov, number NCT01261312. Findings Between Aug 24, 2012, and Sept 15, 2014, 107 patients were enrolled: 54 on the 5-day schedule (26 randomly assigned to 60 mg/m 2 and 28 to 90 mg/m 2 ) and 53 were assigned to the 10-day schedule. Median age was 77 years (range 62–92), and median follow-up was 953 days (IQR 721–1040). All treated patients were assessable for a response. The number of patients who achieved a composite complete response did not differ between dose groups or schedules (13 [54%, 95% CI 32·8–74·4] with 60 mg/m 2 on the 5-day schedule; 16 [59%; 38·8–77·6] with 90 mg/m 2 on the 5-day schedule; and 26 [50%, 35·8–64·2] with 60 mg/m 2 on the 10-day schedule). The most frequent grade 3 or worse adverse events, regardless of relationship to treatment, were febrile neutropenia (31 [61%] of 51 patients on the 5-day schedule vs 36 [69%] of 52 patients on the 10-day schedule), thrombocytopenia (25 [49%] vs 22 [42%]), neutropenia (20 [39%] vs 18 [35%]), pneumonia (15 [29%] vs 19 [37%]), anaemia (15 [29%] vs 12 [23%]), and sepsis (eight [16%] vs 14 [27%]). The most common serious adverse events, regardless of relationship to treatment, for the 5-day and 10-day schedules, respectively, were febrile neutropenia (27 [53%] vs 25 [48%]), pneumonia (14 [27%] vs 16 [31%]), and sepsis (eight [16%] vs 14 [27%]). 23 (22%) patients died because of adverse events (mainly from sepsis, eight [8%]; and pneumonia, five [5%]); four deaths were from adverse events deemed treatment-related (pneumonia, two [2%]; multiorgan failure, one [1%]; and sepsis, one [1%], all in the 10-day cohort). Interpretation More than half of older treatment-naive patients with acute myeloid leukaemia achieved a composite complete response with guadecitabine at all drug doses and schedules investigated, with tolerable toxicity. The recommended guadecitabine regimen for this population is 60 mg/m 2 in a 5-day schedule. A phase 3 study in this patient population is ongoing (NCT02348489) to assess guadecitabine 60 mg/m 2 in a 5-day schedule versus standard of care. Funding Astex Pharmaceuticals and Stand Up To Cancer.
- Published
- 2017
- Full Text
- View/download PDF
12. Acute Myeloid Leukemia, Version 3.2017, NCCN Clinical Practice Guidelines in Oncology
- Author
-
Frederick R. Appelbaum, Keith W. Pratz, Steven Coutre, Dale L. Bixby, Michael Gary Martin, Steven D. Gore, Melanie Fiorella, William Blum, Richard Stone, Deniz Peker, Martin S. Tallman, Daniel A. Pollyea, Farhad Ravandi, Camille N. Abboud, Vijaya Raj Bhatt, Jeffrey E. Lancet, Rebecca L. Olin, Matthew J. Wieduwilt, Stephen A. Strickland, Lori J. Maness, Daniel A. Arber, James M. Foran, Aric C. Hall, Guido Marcucci, Patricia Kropf, Eunice S. Wang, Paul J. Shami, Amir T. Fathi, Kristina M. Gregory, Ndiya Ogba, Jessica K. Altman, Marcos de Lima, Joseph O. Moore, and Margaret R. O'Donnell
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,MEDLINE ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Disease management (health) ,neoplasms ,Acute leukemia ,business.industry ,Age Factors ,Disease Management ,Myeloid leukemia ,medicine.disease ,Clinical Practice ,Transplantation ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,business - Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia among adults and accounts for the largest number of annual deaths due to leukemias in the United States. Recent advances have resulted in an expansion of treatment options for AML, especially concerning targeted therapies and low-intensity regimens. This portion of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for AML focuses on the management of AML and provides recommendations on the workup, diagnostic evaluation and treatment options for younger (age
- Published
- 2017
- Full Text
- View/download PDF
13. Splenic irradiation for splenomegaly: A systematic review
- Author
-
Graeme R. Williams, Shelly B. Hayes, Patricia Kropf, Stefan K. Barta, Nicholas G. Zaorsky, Nestor F. Esnaola, and Joshua E. Meyer
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Gastroenterology ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Radiology, Nuclear Medicine and imaging ,Myelofibrosis ,Cytopenia ,Leukopenia ,business.industry ,General Medicine ,medicine.disease ,Pancytopenia ,Surgery ,Radiation therapy ,Regimen ,Leukemia ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,medicine.symptom ,business - Abstract
Splenic irradiation (SI) is a palliative treatment option for symptomatic splenomegaly (i.e. for pain, early satiety, pancytopenia from sequestration) secondary to hematologic malignancies and disorders. The purpose of the current article is to review the literature on SI for hematologic malignancies and disorders, including: (1) patient selection and optimal technique; (2) efficacy of SI; and (3) toxicities of SI. PICOS/PRISMA methods are used to select 27 articles including 766 courses of SI for 486 patients from 1960 to 2016. The most common cancers treated included chronic lymphocytic leukemia and myeloproliferative disorders; the most common regimen was 10Gy in 1Gy fractions over two weeks, and 27% of patients received retreatment. A partial or complete response (for symptoms, lab abnormalities) was obtained in 85-90% of treated patients, and 30% were retreated within 6-12months. There was no correlation between biologically equivalent dose of radiation therapy and response duration, pain relief, spleen reduction, or cytopenia improvement (r2 all
- Published
- 2017
- Full Text
- View/download PDF
14. Genomic and epigenomic predictors of response to guadecitabine in relapsed/refractory acute myelogenous leukemia
- Author
-
Jean Pierre J. Issa, Hagop M. Kantarjian, Jaroslav Jelinek, Henry C. Fung, Mohammad Azab, Patricia Kropf, Woonbok Chung, Xiang Yao Su, Gail J. Roboz, and Andrew Kelly
- Subjects
0301 basic medicine ,Oncology ,Neuroblastoma RAS viral oncogene homolog ,Male ,medicine.disease_cause ,0302 clinical medicine ,AML ,CDKN2B ,Genetics (clinical) ,Aged, 80 and over ,Myeloid leukemia ,Genomics ,Guadecitabine ,Middle Aged ,3. Good health ,Leukemia ,Leukemia, Myeloid, Acute ,Treatment Outcome ,030220 oncology & carcinogenesis ,DNA methylation ,Azacitidine ,Female ,KRAS ,Mutations ,Adult ,medicine.medical_specialty ,DNA methyltransferase ,03 medical and health sciences ,Young Adult ,DNA methylation inhibitor ,Internal medicine ,Genetics ,medicine ,Biomarkers, Tumor ,Humans ,Molecular Biology ,Survival rate ,Aged ,business.industry ,Research ,DNA Methylation ,medicine.disease ,Survival Analysis ,030104 developmental biology ,Drug resistance ,Mutation ,Gene expression ,Neoplasm Recurrence, Local ,business ,Developmental Biology - Abstract
Background Guadecitabine is a novel DNA methyltransferase (DNMT) inhibitor with improved pharmacokinetics and clinical activity in a subset of patients with relapsed/refractory acute myeloid leukemia (r/r AML), but identification of this subset remains difficult. Methods To search for biomarkers of response, we measured genome-wide DNA methylation, mutations of 54 genes, and expression of a panel of 7 genes in pre-treatment samples from 128 patients treated at therapeutic doses in a phase I/II study. Results Response rate to guadecitabine was 17% (2 complete remission (CR), 3 CR with incomplete blood count recovery (CRi), or CR with incomplete platelets recovery (CRp)) in the phase I component and 23% (14 CR, 9 CRi/CRp) in phase II. There were no strong mutation or methylation predictors of response. Gene expression clustering defined a subset of patients (~ 20%) that had (i) high DNMT3B and low CDKN2B, CTCF, and CDA expression; (ii) enrichment for KRAS/NRAS mutations; (iii) frequent CpG island hypermethylation; (iv) low long interspersed nuclear element 1 (LINE-1) hypomethylation after treatment; and (v) resistance to guadecitabine in both phase I (response rate 0% vs. 33%, p = 0.07) and phase II components of the study (response rate 5% vs. 30%, p = 0.02). Multivariate analysis identified peripheral blood (PB) blasts and hemoglobin as predictors of response and cytogenetics, gene expression, RAS mutations, and hemoglobin as predictors of survival. Conclusions A subset of patients (~ 20%) with r/r AML is unlikely to benefit from guadecitabine as a single agent. In the remaining 80%, guadecitabine is a viable option with a median survival of 8 months and a 2-year survival rate of 21%. Trial registration NCT01261312. Electronic supplementary material The online version of this article (10.1186/s13148-019-0704-3) contains supplementary material, which is available to authorized users.
- Published
- 2019
15. Chimeric Antigen Receptor T-Cell Therapy
- Author
-
Jeff J. Reitan, Stephanie Farnia, Elizabeth J. Shpall, Robert W. Carlson, Lauren Gurschick, Craig S. Sauter, Terry J. Fry, Bijal D. Shah, Patricia Kropf, Steven T. Rosen, Ndiya Ogba, Rebecca A. Gardner, Elizabeth Lihua Budde, Christine E. Brown, Nicole M. Arwood, Nancy L. Bartlett, Patrick A. Brown, Morgan Garber, and Mara Bloom
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,T-Lymphocytes ,Advisory Committees ,Drug resistance ,Cancer Care Facilities ,Medical Oncology ,Immunotherapy, Adoptive ,Transplantation, Autologous ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Neoplasms ,medicine ,Humans ,Stage (cooking) ,Societies, Medical ,Receptors, Chimeric Antigen ,business.industry ,Health Policy ,Cancer ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,United States ,Lymphoma ,Transplantation ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Chimeric Antigen Receptor T-Cell Therapy ,Interdisciplinary Communication ,Neoplasm Recurrence, Local ,business - Abstract
Patients with relapsed or refractory (R/R) cancers have a poor prognosis and limited treatment options. The recent approval of 2 chimeric antigen receptor (CAR) autologous T-cell products for R/R B-cell acute lymphoblastic leukemia and non-Hodgkin's lymphoma treatment is setting the stage for what is possible in other diseases. However, there are important factors that must be considered, including patient selection, toxicity management, and costs associated with CAR T-cell therapy. To begin to address these issues, NCCN organized a task force consisting of a multidisciplinary panel of experts in oncology, cancer center administration, and health policy, which met for the first time in March 2018. This report describes the current state of CAR T-cell therapy and future strategies that should be considered as the application of this novel immunotherapy expands and evolves.
- Published
- 2018
16. S879 RESULTS OF ASTRAL-1 STUDY, A PHASE 3 RANDOMIZED TRIAL OF GUADECITABINE (G) VS TREATMENT CHOICE (TC) IN TREATMENT NAÏVE ACUTE MYELOID LEUKEMIA (TN-AML) NOT ELIGIBLE FOR INTENSIVE CHEMOTHERAPY (IC)
- Author
-
Patricia Kropf, X. Thomas, P. Fenaux, J. Brandwein, Y. H. Min, J. Krauter, J. Novak, H. Kantarjian, J. Mayer, G.J. Roboz, Mohammad Azab, Hartmut Döhner, Wiesław Wiktor Jędrzejczak, Judit Demeter, K. Döhner, Y. Miyazaki, L. Gercheva-Kyuchukova, M. Gobbi, J.-P. Issa, Elizabeth A. Griffiths, T. Robak, K. Yee, S.-P. Yeh, and M. Ojeda-Uribe
- Subjects
Oncology ,Therapy naive ,medicine.medical_specialty ,Randomized controlled trial ,business.industry ,law ,Internal medicine ,medicine ,Myeloid leukemia ,Hematology ,Intensive chemotherapy ,business ,law.invention - Published
- 2019
- Full Text
- View/download PDF
17. DNA Hypomethylating Drugs in Cancer Therapy
- Author
-
Takahiro Sato, Jean Pierre J. Issa, and Patricia Kropf
- Subjects
0301 basic medicine ,Antimetabolites, Antineoplastic ,medicine.medical_treatment ,Azacitidine ,Decitabine ,Biology ,Pharmacology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Microenvironment ,Humans ,Clinical Trials as Topic ,Guadecitabine ,Cancer ,Immunotherapy ,medicine.disease ,DNA Demethylation ,030104 developmental biology ,DNA demethylation ,DNA methylation ,Cancer cell ,Cancer research ,medicine.drug ,Perspectives - Abstract
Aberrant DNA methylation is a critically important modification in cancer cells, which, through promoter and enhancer DNA methylation changes, use this mechanism to activate oncogenes and silence of tumor-suppressor genes. Targeting DNA methylation in cancer using DNA hypomethylating drugs reprograms tumor cells to a more normal-like state by affecting multiple pathways, and also sensitizes these cells to chemotherapy and immunotherapy. The first generation hypomethylating drugs azacitidine and decitabine are routinely used for the treatment of myeloid leukemias and a next-generation drug (guadecitabine) is currently in clinical trials. This review will summarize preclinical and clinical data on DNA hypomethylating drugs as a cancer therapy.
- Published
- 2017
18. Results from a Global Randomized Phase 3 Study of Guadecitabine (G) Vs Treatment Choice (TC) in 815 Patients with Treatment Naïve (TN) AML Unfit for Intensive Chemotherapy (IC) ASTRAL-1 Study: Analysis By Number of Cycles
- Author
-
Marco Gobbi, Jan Novák, Pierre Fenaux, Hagop M. Kantarjian, Su-Peng Yeh, Xavier Thomas, Judit Demeter, Yasushi Miyazaki, Karen W.L. Yee, Joseph Brandwein, Liana Gercheva, Yong Hao, Mario Ojeda-Uribe, Patricia Kropf, Hartmut Döhner, Jean Pierre J. Issa, Tadeusz Robak, Jürgen Krauter, Gail J. Roboz, Jiri Mayer, Yoo Hong Min, Elizabeth A. Griffiths, Wiesław Wiktor Jędrzejczak, and Mohammad Azab
- Subjects
Oncology ,medicine.medical_specialty ,Intention-to-treat analysis ,Surrogate endpoint ,business.industry ,medicine.medical_treatment ,Immunology ,Decitabine ,Phases of clinical research ,Epley maneuver ,Cell Biology ,Hematology ,Biochemistry ,Chemotherapy regimen ,Internal medicine ,Cytarabine ,medicine ,Adverse effect ,business ,medicine.drug - Abstract
Background: Guadecitabine (G) is a next generation subcutaneous (SC) hypomethylating agent (HMA) resistant to degradation by cytidine deaminase which results in prolonged in vivo exposure to the active metabolite decitabine. We conducted a large global randomized phase 3 study of G vs Treatment Choice (TC) with azacitidine (AZA), decitabine (DEC), or low dose Ara-C (LDAC) in 815 TN AML patients unfit for IC (ASTRAL-1 study). The primary ITT results were previously presented (Fenaux et al, EHA abstract S879, 2019). Clinical guidelines for single agent HMAs recommend a minimum of 4 to 6 treatment cycles for maximum benefit. We describe here the results of the study based on number of treatment cycles administered. M ethods: TN-AML patients ineligible for IC due to age ≥ 75 y, or coexisting morbidities, or ECOG PS 2-3 were randomized 1:1 to either G (60 mg/m2/d SC for 5-days Q28 days) or a preselected TC of AZA, DEC, or LDAC at their standard dose/schedule. AML diagnosis and response status were assessed by an independent central pathologist blinded to randomization assignment. Complete response (CR) and overall survival (OS) were co-primary endpoints. We analyzed patients' characteristics, number of treatment cycles, reasons for treatment discontinuation, CR, and OS including analyses by number of cycles received including prospective subgroups, and OS analyses of responders and non-responders. Results: 815 patients were randomized to G (408) or TC (407). Preselected TCs prior to randomization were DEC (43%), AZA (42%), and LDAC (15%). Baseline variables were well balanced across the 2 treatment arms. For G vs TC respectively, age ≥75 y in 62% vs 62.4%, PS 2-3 in 50.5% vs 50.4% (including 10.8% vs 8.8% PS 3), and poor risk cytogenetics in 34.3% vs 34.6%. Most patients were assigned to an HMA at randomization (759, 93%) with only 56 patients (7%) randomized to receive LDAC. Both CR (19.4% for G and 17.4% for TC), and OS Hazard Ratio (0.97; 95% CI 0.83-1.14) were similar and not significantly different between G and TC. Many patients in both arms did not receive the recommended minimum of 4 cycles (42.4% vs 40.8% for G vs TC respectively), or 6 cycles (54.2% vs 53.8% for G vs TC). The proportions were well balanced between the 2 treatment arms. Characteristics of patients who received at least 4 or 6 cycles were also well balanced between the 2 treatment arms for age, PS 2-3, secondary AML, poor risk cytogenetics, BM blasts >30%, and proliferative AML (total white cell count ≥20,000/uL). The primary reasons and proportions for treatment discontinuation were similar for the 2 treatments arms. For patients with Summary/Conclusions: In a large global 815-patient randomized study of G vs TC composed mainly of first generation HMAs, G was at least as effective as TC based on the primary ITT analysis of CR and the narrow 95% CI of OS HR (0.83-1.14). Analyses of patients by number of treatment cycles showed that those who received at least 4 or 6 cycles achieved longer OS in G vs TC with the largest benefit in those who received at least 6 cycles. The benefit was observed in all subgroups, and in both responders and non-responders. Treatment with single agent guadecitabine should continue as long as the patient can still benefit and for at least 6 cycles to gain the maximum survival benefit. Disclosures Roboz: Trovagene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sandoz: Consultancy, Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Actinium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amphivena: Consultancy, Membership on an entity's Board of Directors or advisory committees; Argenx: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astex: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celltrion: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Eisai: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; MEI Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Orsenix: Consultancy, Membership on an entity's Board of Directors or advisory committees; Otsuka: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Roche/Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees. Döhner:Celgene, Novartis, Sunesis: Honoraria, Research Funding; AbbVie, Agios, Amgen, Astellas, Astex, Celator, Janssen, Jazz, Seattle Genetics: Consultancy, Honoraria; AROG, Bristol Myers Squibb, Pfizer: Research Funding. Mayer:AOP Orphan Pharmaceuticals AG: Research Funding. Krauter:Pfizer: Honoraria. Robak:Takeda: Consultancy, Research Funding; UCB: Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Other: Travel grant, Research Funding; Amgen: Consultancy, Other: Travel grant; Roche: Consultancy, Other: Travel grant, Research Funding; Abbvie: Consultancy, Honoraria, Other: Travel grant, Research Funding; Gilead: Consultancy, Research Funding; BeiGene: Consultancy, Research Funding; Acerta: Research Funding; Morphosys AG: Research Funding. Kantarjian:Agios: Honoraria, Research Funding; Astex: Research Funding; Jazz Pharma: Research Funding; Amgen: Honoraria, Research Funding; BMS: Research Funding; Novartis: Research Funding; Actinium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Immunogen: Research Funding; AbbVie: Honoraria, Research Funding; Takeda: Honoraria; Cyclacel: Research Funding; Pfizer: Honoraria, Research Funding; Daiichi-Sankyo: Research Funding; Ariad: Research Funding. Novak:Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Roche: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel,Accommodations; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Other: Travel,Accommodations. Jedrzejczak:Takeda: Consultancy; Amgen: Consultancy, Other: travel support for hematology meetings; Celgene: Other: travel support for hematology meetings; Novartis: Research Funding; Roche: Other: travel support for hematology meetings. Thomas:PFIZER: Honoraria; ABBVIE: Honoraria; DAICHI: Honoraria; INCYTE: Honoraria. Miyazaki:Chugai: Research Funding; Otsuka: Honoraria; Novartis: Honoraria; Nippon-Shinyaku: Honoraria; Dainippon-Sumitomo: Honoraria; Kyowa-Kirin: Honoraria. Brandwein:Jazz Pharma: Consultancy, Honoraria; Otsuka: Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Roche: Research Funding; Novartis: Consultancy, Honoraria. Demeter:Angelini: Other: Advisory Board; Pfizer: Other: Advisory Board; Novartis: Other: Advisory Board; Bristol Myers Squibb: Other: Advisory Board; Amicus: Other: Advisory Board; Amgen: Other: Advisory Board; Roche: Other: Advisory Board. Griffiths:Astex Phramaceuticals/Otsuka Pharmaceuticals: Consultancy, Research Funding; Persimmune: Consultancy; Persimmune: Consultancy; Genentech, Inc.: Research Funding; Appelis Pharmaceuticals: Other: PI on a clinical trial; New Link Genetics: Consultancy; Novartis Inc.: Consultancy; Novartis Inc.: Consultancy; Onconova Therapeutics: Other: PI on a clinical trial; Partner Therapeutics: Consultancy; Appelis Pharmaceuticals: Other: PI on a clinical trial; Boston Scientific: Consultancy; Boston Scientific: Consultancy; Genentech, Inc.: Research Funding; Abbvie, Inc.: Consultancy; Celgene, Inc: Consultancy, Research Funding; Celgene, Inc: Consultancy, Research Funding; New Link Genetics: Consultancy; Onconova Therapeutics: Other: PI on a clinical trial; Partner Therapeutics: Consultancy; Astex Phramaceuticals/Otsuka Pharmaceuticals: Consultancy, Research Funding; Abbvie, Inc.: Consultancy, PI on a clinical trial. Yee:Agensys, Astex, Hoffman La Roche, MedImmune, Merck, Millenium, Roche/Genentech: Research Funding; Novartis, Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Astellas, Celgene, Otsuka, Shire, Takeda: Membership on an entity's Board of Directors or advisory committees. Hao:Astex Pharmaceuticals, Inc.: Employment. Azab:Astex Pharmaceuticals, Inc.: Employment. Fenaux:Celgene Corporation: Honoraria, Research Funding; Aprea: Research Funding; Astex: Honoraria, Research Funding; Jazz: Honoraria, Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
19. Chronic Myelogenous Leukemia, Version 1.2014
- Author
-
Madan Jagasia, Kristina M. Gregory, Arnel Pallera, David S. Snyder, Jerald P. Radich, Jason Gotlib, Javier Pinilla-Ibarz, Ellin Berman, Susan O'Brien, Daniel J. DeAngelo, Jessica K. Altman, Patricia Kropf, Neil P. Shah, Steven M. Devine, Hema Sundar, B. Douglas Smith, Joseph O. Moore, Mojtaba Akhtari, Vishnu Reddy, Meir Wetzler, Amir T. Fathi, Michael W. Deininger, and Camille N. Abboud
- Subjects
Oncology ,medicine.medical_specialty ,medicine.drug_class ,International scale ,Fusion Proteins, bcr-abl ,Antineoplastic Agents ,Article ,Tyrosine-kinase inhibitor ,Myelogenous ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Protein Kinase Inhibitors ,business.industry ,Imatinib ,Prognosis ,medicine.disease ,Surgery ,Dasatinib ,Leukemia ,Nilotinib ,business ,Chronic myelogenous leukemia ,medicine.drug - Abstract
The 2014 NCCN Clinical Practice Guidelines in Oncology for Chronic Myelogenous Leukemia recommend quantitative reverse-transcription polymerase chain reaction (QPCR) standardized to International Scale (IS) as the preferred method for monitoring molecular response to tyrosine kinase inhibitor (TKI) therapy. A BCR-ABL1 transcript level of 10% or less (IS) is now included as the response milestone at 3 and 6 months. Change of therapy to an alternate TKI is recommended for patients with BCR-ABL1 transcript levels greater than 10% (IS) at 3 months after primary treatment with imatinib. Continuing the same dose of TKI or switching to an alternate TKI are options for patients with BCR-ABL1 transcript levels greater than 10% (IS) at 3 months after primary treatment with dasatinib or nilotinib. The guidelines recommend 6-month evaluation with QPCR (IS) for patients with BCR-ABL1 transcript levels greater than 10% at 3 months. Monitoring with QPCR (IS) every 3 months is recommended for all patients, including those who meet response milestones at 3, 6, 12, and 18 months (BCR-ABL1 transcript level ≤10% [IS] at 3 and 6 months, complete cytogenetic response at 12 and 18 months).
- Published
- 2013
- Full Text
- View/download PDF
20. Myeloproliferative Neoplasms, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology
- Author
-
Kristina M. Gregory, Sanjay R. Mohan, Raajit K. Rampal, Brady L. Stein, Lindsay A.M. Rein, Bart L. Scott, Ivana Gojo, Rebecca B. Klisovic, Nikolai A. Podoltsev, Daniel A. Pollyea, Srdan Verstovsek, Krishna Gundabolu, Stephen T. Oh, Ravi Bhatia, Michael W. Deininger, Eric Padron, Aaron T. Gerds, Mary Anne Bergman, Patricia Kropf, Hema Sundar, Gabriela S. Hobbs, Ruben A. Mesa, David S. Snyder, Jason Gotlib, Martha Wadleigh, Catriona Jamieson, and Eunice S. Wang
- Subjects
Oncology ,medicine.medical_specialty ,Philadelphia chromosome ,Medical Oncology ,Risk Assessment ,03 medical and health sciences ,0302 clinical medicine ,Polycythemia vera ,Quality of life ,hemic and lymphatic diseases ,Internal medicine ,Prevalence ,Medicine ,Humans ,Philadelphia Chromosome ,Myelofibrosis ,Polycythemia Vera ,business.industry ,Essential thrombocythemia ,Janus Kinase 2 ,medicine.disease ,Clinical Practice ,Primary Myelofibrosis ,030220 oncology & carcinogenesis ,Risk stratification ,Mutation ,Quality of Life ,business ,Calreticulin ,Receptors, Thrombopoietin ,030215 immunology ,Signal Transduction ,Thrombocythemia, Essential - Abstract
Myelofibrosis (MF), polycythemia vera (PV), and essential thrombocythemia (ET) are a group of heterogeneous disorders of the hematopoietic system collectively known as Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). The diagnosis and the management of patients with MPNs have evolved since the identification of mutations that activate the JAK pathway (JAK2, CALR, and MPL mutations) and the development of targeted therapies has resulted in significant improvements in disease-related symptoms and quality of life. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnostic workup of MPN (MF, PV, and ET), risk stratification, treatment, and supportive care strategies for the management of MF.
- Published
- 2016
21. NCCN Guidelines Insights: Chronic Myeloid Leukemia, Version 1.2017
- Author
-
Kristina M. Gregory, Madan Jagasia, Hagop M. Kantarjian, Enkhtsetseg Purev, Joseph O. Moore, B. Douglas Smith, Daniel J. DeAngelo, Patricia Kropf, Michal G. Rose, Vishnu Reddy, Kendra Sweet, Raoul Tibes, Camille N. Abboud, Neil P. Shah, Hema Sundar, Evelena P. Ontiveros, Gabriela S. Hobbs, R. Tanner Hagelstrom, Arnel Pallera, Michael W. Deininger, David T. Yang, Ellin Berman, Bhavana Bhatnagar, Peter T. Curtin, David S. Snyder, Jason Gotlib, Jerald P. Radich, Albert Thomas Quiery, Jessica K. Altman, and Leland Metheny
- Subjects
0301 basic medicine ,Adult ,Pediatrics ,medicine.medical_specialty ,Myeloid ,media_common.quotation_subject ,MEDLINE ,Fertility ,Reproductive age ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,hemic and lymphatic diseases ,Medicine ,Humans ,Child ,neoplasms ,Protein Kinase Inhibitors ,media_common ,Aged ,Evidence-Based Medicine ,business.industry ,Myeloid leukemia ,Abnormalities, Drug-Induced ,medicine.disease ,Prognosis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Withholding Treatment ,030220 oncology & carcinogenesis ,Leukemia, Myeloid, Chronic-Phase ,Practice Guidelines as Topic ,Female ,business ,Pregnancy Complications, Neoplastic ,Pediatric population - Abstract
The NCCN Guidelines for Chronic Myeloid Leukemia (CML) provide recommendations for the management of chronic-phase and advanced-phase CML in adult patients. The median age of disease onset is 67 years. However, because CML occurs in all age groups, clinical care teams should be prepared to address issues relating to fertility and pregnancy with patients who are of reproductive age at the time of diagnosis. CML is relatively rare in children and there are no evidence-based recommendations for the management of CML in pediatric population. These NCCN Guidelines Insights discuss special considerations for the management of CML during pregnancy and for the management of CML in the pediatric population.
- Published
- 2016
22. Randomized Phase 2 Study of Guadecitabine in Patients with HMA-Naive Higher Risk Myelodysplastic Syndromes (MDS) or Chronic Myelomonocytic Leukemia (CMML)
- Author
-
Casey O'Connell, Hagop M. Kantarjian, Michael R. Savona, Raoul Tibes, Sue Naim, G. Garcia-Manero, Patricia Kropf, Gail J. Roboz, Mohammad Azab, Wendy Stock, Scott D. Lunin, J.-P. Issa, Yong Hao, Karen W.L. Yee, Jesus G. Berdeja, and Katherine Walsh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Myelodysplastic syndromes ,Chronic myelomonocytic leukemia ,Phases of clinical research ,Hematology ,medicine.disease ,Internal medicine ,medicine ,In patient ,business - Published
- 2017
- Full Text
- View/download PDF
23. CMV and BK Virus Reactivation in Post Transplant Cyclophosphamide (PTCY) after Allogeneic MUD, MRD and Haploidentical Transplant
- Author
-
Carlyn Rose C. Tan, Patricia Kropf, Trent P Wang, Stefan K. Barta, John Ulicny, Matthew Sochat, Henry C. Fung, Nasheed Hossain, and Philip A. Pancari
- Subjects
0301 basic medicine ,Transplantation ,business.industry ,Post transplant cyclophosphamide ,Hematology ,medicine.disease_cause ,Virology ,BK virus ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,business - Published
- 2017
- Full Text
- View/download PDF
24. An Evaluation of Mobilization Efficiency, Efficacy and Toxicity among 3 Different Approaches: Chemomobilization, Chemomobilization with Preemptive Plerixafor Use and Plerixafor with G-CSF as Stem Cell Mobilization (SCM) Strategies in Lymphoid Malignancies
- Author
-
Patricia Kropf, Carlyn Rose C. Tan, John Ulicny, Stefan K. Barta, Philip A. Pancari, Henry C. Fung, and Trent P Wang
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,Mobilization ,Stem cell mobilization ,business.industry ,Plerixafor ,Internal medicine ,Toxicity ,medicine ,Hematology ,business ,medicine.drug - Published
- 2017
- Full Text
- View/download PDF
25. Long Term Results of a Randomized Phase 2 Dose-Response Study of Guadecitabine, a Novel Subcutaneous (SC) Hypomethylating Agent (HMA), in 102 Patients with Intermediate or High Risk Myelodysplastic Syndromes (MDS) or Chronic Myelomonocytic Leukemia (CMML)
- Author
-
Katherine Walsh, Yong Hao, Mohammad Azab, Hagop M. Kantarjian, Nikolai A. Podoltsev, Patricia Kropf, Sue Naim, Guillermo Garcia-Manero, Jesus G. Berdeja, Naval Daver, Ellen K. Ritchie, Karen W.L. Yee, Joseph R. Mace, Wendy Stock, Jean Pierre J. Issa, Raoul Tibes, Elias Jabbour, Casey O'Connell, Gail J. Roboz, Elizabeth A. Griffiths, Michael R. Savona, Todd L. Rosenblat, and Scott D. Lunin
- Subjects
medicine.medical_specialty ,education.field_of_study ,business.industry ,Myelodysplastic syndromes ,Immunology ,Population ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,Long term results ,medicine.disease ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Hypomethylating agent ,Median follow-up ,030220 oncology & carcinogenesis ,Internal medicine ,Maximum tolerated dose ,Medicine ,business ,education ,Febrile neutropenia ,030215 immunology - Abstract
Background: Guadecitabine SC (SGI-110) is a dinucleotide next generation HMA resistant to degradation by cytidine deaminase resulting in extended in vivo exposure to its active metabolite decitabine. A Phase 1 established 60 mg/m2 QDx5 as the biologically effective dose (BED), and 90 mg/m2 QDx5 as the Maximum tolerated dose (MTD) in MDS patients given in 28-day cycles (Issa et al, 2015, Lancet Oncology). Phase 2 is conducted to evaluate dose response between the BED and MTD in both untreated MDS patients, and patients previously treated with other HMAs. M ethods: Int, or HR MDS, and CMML patients who were either treatment-naïve (TN) or relapsed/refractory to other HMAs (r/r) were randomized to either 60 mg/m2 or 90 mg/m2 QDx5 every 28 days. Efficacy was evaluated by the clinical responses of CR, PR, marrow CR (mCR), and Hematological Improvement (HI) based on the International Working Group Criteria 2006, as well as transfusion-independence, and overall survival (OS). Adverse events (AEs) were graded by the CTCAE v4 criteria. Results: The study completed target enrolment with 102 patients: 53 r/r MDS, and 49 TN MDS. Fifty three patients were randomized to 60 mg/m2 and 49 patients to 90 mg/m2 QDx5 with a median follow up of 3.2 years (IQR 2.8-3.5 years). Median age was 72 and 71 years for r/r and TN MDS respectively. Most baseline patient characteristics were well balanced between the 2 treatment dose groups except that more CMML patients were randomized to the 60 mg/m2 group (28%) vs. 14% in the 90 mg/m2 group, and more patients with baseline BM blasts >5% were in the 90 mg/m2 group (67%) vs. 38% in the 60 mg/m2 group. Most patients were RBC transfusion-dependent at baseline (57%). In the r/r MDS cohort, most patients (58%) received their last HMA treatment Median number of treatment cycles was 5 for both r/r and TN MDS (range 1-37 in r/r MDS and 1-49 in TN MDS). In the TN MDS cohort CR was achieved in 11 (22%) of patients with no major difference between the 2 dose groups (19% in the 60 mg/m2 group vs 27% in the 90 mg/m2 group). Overall CR+mCR was achieved in 18 patients (37%) in TN MDS patients and median OS was 23.4 months. In the r/r MDS cohort, CR was achieved in 4% of patients in each of the 2 dose groups. Overall CR+mCR in the r/r MDS cohort was achieved in 17 patients (32%), with a median duration of response of 7.9 months, and median OS of 11.7 months. No significant difference in response or OS between the 2 dose groups was observed. In patients who were RBC transfusion-dependent at baseline, transfusion independence for at least 8 weeks was achieved in 42% of TN MDS, and 15% in r/r MDS patients. In the overall population of 102 TN and r/r MDS patients there were no major differences in OS based on DNMT3A or TET2 mutation status while patients with TP53 mutations had worse median OS (7.4 months) compared to those without TP53 mutations (22.6 months). Other baseline prognostic factors for worse OS were BM blasts >5%; RBC transfusion-dependence; IPSS High Risk; and ECOG Performance Status of >1. Overall incidence of Grade ≥3 AEs regardless of relationship to treatment was reported in 83 vs. 96% for 60 and 90 mg/m2 dose groups respectively. There was a slightly higher but non-significant difference in Grade ≥3 thrombocytopenia (57 vs 41.5%); neutropenia (51 vs 39.6%); febrile neutropenia (43% vs 32%); and pneumonia (32.7 vs. 26.4%) for the 90 mg/m2 compared to 60 mg/m2 dose group. Early 30, 60, and 90-day all-cause mortality was observed in 0, 3.7%, and 5.7% in the 60 mg/m2 dose group respectively; and in 2%, 4%, and 12% in the 90 mg/m2 dose group respectively. Conclusions: Guadecitabine at both dose groups is a well-tolerated novel HMA with clinical activity in the treatment of both TN and r/r Int and HR MDS, and CMML patients. In TN MDS patient CR rate of 22% and median OS of 23.4 months compare well with first generation HMA efficacy (Fenaux et al, 2009, Lancet Oncology). Activity in r/r MDS who previously failed prior HMAs is particularly promising (CR+mCR in 32% of patients with median duration of response and overall survival of almost 8 and 12 months respectively). A phase 3 trial (ASTRAL-3) of guadecitabine vs Physician Treatment Choice in r/r MDS and CMML patients previously treated with azacitidine or decitabine is actively enrolling (ClinicalTrials.gov ID: NCT02907359). Disclosures Ritchie: NS Pharma: Research Funding; Bristol-Myers Squibb: Research Funding; Novartis: Consultancy, Other: Travel, Accommodations, Expenses, Research Funding, Speakers Bureau; Incyte: Consultancy, Speakers Bureau; Celgene: Consultancy, Other: Travel, Accommodations, Expenses, Speakers Bureau; Pfizer: Consultancy, Research Funding; ARIAD Pharmaceuticals: Speakers Bureau; Astellas Pharma: Research Funding. Savona:Boehringer Ingelheim: Consultancy; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding. Kropf:Celegene: Consultancy; Takeda: Consultancy. Daver:Pfizer: Consultancy; Sunesis: Consultancy; Pfizer: Research Funding; ImmunoGen: Consultancy; ARIAD: Research Funding; Novartis: Research Funding; Incyte: Research Funding; Otsuka: Consultancy; Novartis: Consultancy; Karyopharm: Consultancy; Alexion: Consultancy; Incyte: Consultancy; Karyopharm: Research Funding; BMS: Research Funding; Kiromic: Research Funding; Daiichi-Sankyo: Research Funding; Sunesis: Research Funding. Jabbour:Pfizer: Consultancy, Research Funding; Novartis: Research Funding; Abbvie: Research Funding; Takeda: Consultancy, Research Funding; Bristol-Myers Squibb: Consultancy, Research Funding. Yee:Celgene, Novartis, Otsuka: Membership on an entity's Board of Directors or advisory committees; Agensys, Astex, GSK, Onconova, Genentech/Roche: Research Funding. Stock:Jazz Pharmaceuticals: Consultancy. Griffiths:Alexion Inc.: Honoraria, Research Funding; Astex/Otsuka Pharmaceuticals: Honoraria, Research Funding; Celgene, Inc: Honoraria, Research Funding; Novartis, Inc.: Research Funding; Pfizer, Inc.: Research Funding. Podoltsev:Astex Pharmaceuticals: Research Funding; LAM Therapeutics: Research Funding; Sunesis Pharmaceuticals: Research Funding; Genentech: Research Funding; Boehringer Ingelheim: Research Funding; Daiichi Snakyo: Research Funding; Astellas Pharma: Research Funding; Pfizer: Research Funding; Celator: Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding. Berdeja:Genentech: Research Funding; Bluebird: Research Funding; Amgen: Research Funding; Novartis: Research Funding; Glenmark: Research Funding; Takeda: Research Funding; Bristol-Myers Squibb: Research Funding; Teva: Research Funding; Sanofi: Research Funding; Poseida Therapeutics, Inc.: Research Funding; Celgene: Research Funding; Janssen: Research Funding. Naim:Astex Pharmaceuticals, Inc.: Employment. Hao:Astex Pharmaceuticals, Inc.: Employment. Azab:Astex Pharmaceuticals, Inc.: Employment. Roboz:Orsenix: Consultancy; Roche/Genentech: Consultancy; Roche/Genentech: Consultancy; Otsuka: Consultancy; Bayer: Consultancy; Pfizer: Consultancy; Jazz Pharmaceuticals: Consultancy; Argenx: Consultancy; Sandoz: Consultancy; Celltrion: Consultancy; Bayer: Consultancy; Orsenix: Consultancy; Pfizer: Consultancy; Astex Pharmaceuticals: Consultancy; Daiichi Sankyo: Consultancy; Sandoz: Consultancy; Daiichi Sankyo: Consultancy; Novartis: Consultancy; Janssen Pharmaceuticals: Consultancy; Cellectis: Research Funding; Eisai: Consultancy; Celgene Corporation: Consultancy; Jazz Pharmaceuticals: Consultancy; Celgene Corporation: Consultancy; Janssen Pharmaceuticals: Consultancy; Eisai: Consultancy; Novartis: Consultancy; Otsuka: Consultancy; Argenx: Consultancy; Celltrion: Consultancy; Astex Pharmaceuticals: Consultancy; Aphivena Therapeutics: Consultancy; AbbVie: Consultancy; Aphivena Therapeutics: Consultancy; AbbVie: Consultancy; Cellectis: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
26. Phase I Results of a Multicenter Clinical Trial Combining Guadecitabine, a DNA Methyltransferase Inhibitor, with Atezolizumab, an Immune Checkpoint Inhibitor, in Patients with Relapsed or Refractory Myelodysplastic Syndrome or Chronic Myelomonocytic Leukemia
- Author
-
Casey O'Connell, Patricia Kropf, Nathan Punwani, Kirsten Grønbæk, Richard Sposto, and Dan Rogers
- Subjects
Oncology ,medicine.medical_specialty ,Leukopenia ,business.industry ,Immunology ,Chronic myelomonocytic leukemia ,Phases of clinical research ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Tolerability ,Atezolizumab ,030220 oncology & carcinogenesis ,Internal medicine ,Clinical endpoint ,Medicine ,030212 general & internal medicine ,medicine.symptom ,business - Abstract
Background: There are no FDA-approved therapies for patients with myelodysplastic syndrome (MDS) in whom hypomethylating agents (HMAs) fail, and mechanisms of resistance are not well-characterized. Preclinical and clinical data suggest that some myeloblasts express PDL-1 and that HMAs can induce expression of PD-1 on T cells, which may result in resistance through immune tolerance. We hypothesized that the addition of the PDL1-inhibitor atezolizumab to guadecitabine, a next generation HMA which has a longer in vivo exposure time, would induce or restore HMA sensitivity in patients with relapsed or refractory (R/R) MDS. Overlapping toxicities were not expected given atezolizumab is not myelosuppressive and guadecitabine does not appear to induce autoimmune or inflammatory conditions. Methods: We are conducting a phase I/II, multicenter clinical trial for adult patients with R/R, intermediate (3+) or high-risk MDS by the revised international scoring system, including chronic myelomonocytic leukemia (CMML). A 3x3 dose escalation design for guadecitabine was used for Phase I, beginning with 30 mg/m2 (Dose level -1) days 1-5 along with a fixed dose of atezolizumab 840mg IV days 8 and 22 of a 28-day cycle. The plan was to escalate to the recommended dose of guadecitabine, 60mg/m2 (Dose Level 1) if no dose limiting toxicities (DLTs) were identified during Dose Level -1. If ≥2/6 DLTs were observed during the Dose Level 1, de-escalation to 45mg/m2 (Dose Level -1.5) would occur. The primary endpoint of phase I was safety and tolerability of the combination. Overall survival (OS) from the on-study date and overall response rates (ORR), based on the 2006 Modified IWG Response Criteria for MDS, were secondary endpoints. Results: Nine patients (5M, 4F, median age 73) with intermediate or higher risk were treated during phase I. Three patients were treated at Dose Level -1 and sustained no DLTs. Similarly, no DLTs were observed among 6 patients treated at Dose Level 1. There were 17 grade 3 or 4 events considered possibly or probably related to the study treatments, the most common of which were: neutropenia (4), thrombocytopenia (4), and leukopenia (4). The median number of treatment cycles was 5 and the treatment duration for each patient is illustrated in Figure 1. Two patients achieved hematologic improvement (HI) and 1 patient achieved CR. Two patients died after coming off of the study (at 4.5 and 9 months respectively) and the median OS has not been reached. Discussion: The combination of guadecitabine at the recommended dose of 60mg/m2 D1-D5 along with atezolizumab 840mg IV d8, 22 was found to be safe with an acceptable toxicity profile in patients with R/R MDS. The ORR was 33% and a phase II study is ongoing. Disclosures Kropf: Celegene: Consultancy; Takeda: Consultancy. Grønbæk:Otsuka Pharma: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Janssen Pharma: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
- Full Text
- View/download PDF
27. Epigenetics and Epigenetic Therapy of Cancer
- Author
-
Omotayo Fasan, Jean Pierre J. Issa, Patricia Kropf, and Patrick Boland
- Subjects
Leukemia ,Histone ,biology ,business.industry ,DNA methylation ,medicine ,Cancer research ,biology.protein ,Cancer ,Epigenetics ,medicine.disease ,business ,Epigenetic therapy - Published
- 2015
- Full Text
- View/download PDF
28. Healthcare utilization and costs associated with tyrosine kinase inhibitor switching in patients with chronic myeloid leukemia
- Author
-
Patricia Kropf, Ashutosh Pathak, Jean Pierre J. Issa, Gisoo Barnes, and Boxiong Tang
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Databases, Factual ,medicine.drug_class ,Antineoplastic Agents ,Tyrosine-kinase inhibitor ,Treatment failure ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,In patient ,Treatment Failure ,Protein Kinase Inhibitors ,Aged ,business.industry ,Drug Substitution ,Myeloid leukemia ,Hematology ,Health Care Costs ,Middle Aged ,Patient Acceptance of Health Care ,nervous system diseases ,Outpatient visits ,Healthcare utilization ,030220 oncology & carcinogenesis ,Immunology ,Cohort ,Retreatment ,Female ,business ,030215 immunology - Abstract
Differences in healthcare utilization and costs were examined in chronic myeloid leukemia (CML) patients experiencing first-, second- and third-line tyrosine kinase inhibitor (TKI) therapy. Three CML cohorts were identified from the Truven Health MarketScan® database: No-Switch Cohort (NSc) = did not switch from first-line; One-Switch Cohort (OSc) = switched from first- to second-line only; Two-Switch Cohort (TSc) = switched to second- and then third-line. A total of 3510 patients were identified (mean = 54%; age = 55.8 years). NSc comprised 81% of the sample, OSc comprised 15% and 4% were in the TSc. First-line utilization/costs were significantly higher in the OSc/TSc compared to the NSc. Second-line hospital/outpatient visits and costs were higher in TSc compared to OSc. TSc experienced a significant cost increase from first- to second-line ($4226.46), twice that of OSc ($2488.03). TKI switching is associated with a substantial increase in healthcare utilization and costs, particularly for patients who switch twice.
- Published
- 2015
29. Donor Lymphocyte Infusion in Hematologic Malignancies--Good to be Fresh?
- Author
-
Michael Garner, Thomas R. Klumpp, Nasheed Hossain, Mangan Kf, Mary Ellen Martin, John Ulicny, Stefan K. Barta, Henry C. Fung, and Patricia Kropf
- Subjects
Cancer Research ,medicine.medical_specialty ,Lymphocyte Transfusion ,medicine.medical_treatment ,Subgroup analysis ,Hematopoietic stem cell transplantation ,Gastroenterology ,Donor lymphocyte infusion ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Retrospective Studies ,business.industry ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Retrospective cohort study ,Hematology ,Chemotherapy regimen ,Tissue Donors ,Surgery ,Transplantation ,Oncology ,030220 oncology & carcinogenesis ,Hematologic Neoplasms ,Neoplasm Recurrence, Local ,business ,030215 immunology ,Follow-Up Studies - Abstract
Background Donor lymphocyte infusion (DLI) has been used with variable success in a variety of hematologic malignancies. Patients and Methods We conducted a retrospective analysis of all patients who were treated with DLI for persistent or relapsed disease at the Temple University Bone Marrow Transplant Unit from July 1, 1993 to December 31, 2013 to evaluate the effect of the type of DLI (fresh vs. cryopreserved) on event-free survival (EFS) and overall survival (OS). Median follow-up was 64.8 months (range, 0.3-142.6 months). Results We found that EFS and OS were similar between patients receiving cryopreserved cells and those receiving fresh DLI (median OS for cryopreserved cells, 0.39 years; median OS for fresh cells, 0.32 years; P = .793; median EFS for cryopreserved cells, 0.410 years; median EFS for fresh cells, 0.420 years; P = .4264). In the setting of relapsed disease, treatment with any chemotherapy regimen before receiving DLI did not significantly impact OS (n = 63; P = .2203) or EFS (n = 40; P = .542). A subgroup analysis limited to patients with acute myeloid leukemia (AML)/myelodysplastic syndrome (MDS) (32 patients) showed that differences in OS and EFS between cryopreserved and fresh DLI approached significance (median OS for cryopreserved cells, 0.34 years; median OS for fresh cells, 0.17 years; P = .16; median EFS for cryopreserved cells, 0.37 years; median EFS for fresh cells, 0.094 years; P = 0.11). Conclusion We conclude that the use of fresh cells versus cryopreserved cells does not have an impact on outcomes, and selected patients can achieve long-term survival with DLI for treatment of relapse after transplantation, although the overall outcomes remain dismal.
- Published
- 2015
30. Healthcare Utilization and Costs Associated With Multiple Switching of Tyrosine Kinase Inhibitor therapy In Patients With Chronic Myeloid Leukemia
- Author
-
Gisoo Barnes, B. Tang, Patricia Kropf, Ashutosh Pathak, and Jean Pierre J. Issa
- Subjects
Healthcare utilization ,business.industry ,medicine.drug_class ,Health Policy ,Cancer research ,Public Health, Environmental and Occupational Health ,Medicine ,Myeloid leukemia ,In patient ,business ,Tyrosine-kinase inhibitor - Published
- 2015
- Full Text
- View/download PDF
31. Historical views, conventional approaches, and evolving management strategies for myeloproliferative neoplasms
- Author
-
Marie Huong Nguyen, Nikolai A. Podoltsev, Srdan Verstovsek, Rami S. Komrokji, Brady L. Stein, Daniel A. Pollyea, Rebecca B. Klisovic, Josef T. Prchal, Catriona Jamieson, Patricia Kropf, Meir Wetzler, Sanjay R. Mohan, Krishna Gundabolu, Neil P. Shah, Ruben A. Mesa, Stephen T. Oh, David S. Snyder, Jason Gotlib, Raul Ribeiro, Moshe Talpaz, Martha Wadleigh, Rita Paschal, Alison R. Moliterno, Murat O. Arcasoy, Bart L. Scott, and Ross L. Levine
- Subjects
Ruxolitinib ,medicine.medical_specialty ,Pathology ,MEDLINE ,Antineoplastic Agents ,Disease pathogenesis ,Article ,Quality of life (healthcare) ,Polycythemia vera ,Essential ,Rare Diseases ,hemic and lymphatic diseases ,Receptors ,medicine ,Humans ,Hydroxyurea ,Thrombocythemia ,Oncology & Carcinogenesis ,Intensive care medicine ,Myelofibrosis ,Polycythemia Vera ,Protein Kinase Inhibitors ,Janus Kinases ,Essential thrombocythemia ,business.industry ,Symptom burden ,Hematopoietic Stem Cell Transplantation ,food and beverages ,Thrombosis ,Hematology ,Janus Kinase 2 ,medicine.disease ,Oncology ,Thrombopoietin ,Primary Myelofibrosis ,Splenomegaly ,business ,Calreticulin ,Receptors, Thrombopoietin ,Thrombocythemia, Essential ,medicine.drug - Abstract
The classical Philadelphia chromosome–negative myeloproliferative neoplasms (MPN), which include essential thrombocythemia, polycythemia vera, and myelofibrosis (MF), are in a new era of molecular diagnosis, ushered in by the identification of the JAK2(V617F) and cMPL mutations in 2005 and 2006, respectively, and the CALR mutations in 2013. Coupled with increased knowledge of disease pathogenesis and refined diagnostic criteria and prognostic scoring systems, a more nuanced appreciation has emerged of the burden of MPN in the United States, including the prevalence, symptom burden, and impact on quality of life. Biological advances in MPN have translated into the rapid development of novel therapeutics, culminating in the approval of the first treatment for MF, the JAK1/JAK2 inhibitor ruxolitinib. However, certain practical aspects of care, such as those regarding diagnosis, prevention of vascular events, choice of cytoreductive agent, and planning for therapies, present challenges for hematologists/oncologists, and are discussed in this article.
- Published
- 2015
32. Progressive Disease after Autologous Stem Cell Transplant (ASCT) for Patients with DLBCL in the Rituximab Era: Outcome Analysis and Evaluation of Prognostic Factors
- Author
-
Henry C. Fung, Mary Ellen Martin, John Ulicny, Nasheed Hossain, Patricia Kropf, and Stefan K. Barta
- Subjects
Oncology ,medicine.medical_specialty ,Transplantation ,business.industry ,Outcome analysis ,Hematology ,medicine.disease ,Internal medicine ,Immunology ,medicine ,Rituximab ,Stem cell ,business ,Progressive disease ,medicine.drug - Published
- 2016
- Full Text
- View/download PDF
33. GVHD Prophylaxis with Post-Transplant High Dose Cyclophosphamide: Impact on Engraftment, Treatment-Related Mortality and Resource Utilization
- Author
-
Mary Ellen Martin, Patricia Kropf, Henry C. Fung, Stefan K. Barta, Nasheed Hossain, and John Ulicny
- Subjects
medicine.medical_specialty ,Transplantation ,business.industry ,Hematology ,Post transplant ,Treatment related mortality ,surgical procedures, operative ,High dose cyclophosphamide ,immune system diseases ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Gvhd prophylaxis ,business ,Resource utilization - Published
- 2016
- Full Text
- View/download PDF
34. Abstract 5382: A DNMT3A-independent hypomethylator phenotype is a unifying epigenetic signature of AML with good risk cytogenetics
- Author
-
Jozef Madzo, Jean Pierre J. Issa, Charly R. Good, Patricia Kropf, Priyanka Madireddi, Andrew Kelly, and Jaroslav Jelinek
- Subjects
Genetics ,Cancer Research ,medicine.medical_specialty ,Oncology ,Cytogenetics ,medicine ,Epigenetics ,Biology ,Signature (topology) ,Phenotype - Abstract
Background: Acute myeloid leukemia (AML) causes the most leukemia-related deaths in the United States, and has frequent mutations in epigenetic regulators, including DNMT3A, IDH, and TET2. Such aberrations have been proposed to transform the epigenetic state in cancer, often involving DNA hypomethylation, however, the genomic specificity, causes, and clinical consequences of such methylation changes in AML remain unclear. Methods: We queried genome-wide CpG methylation using The Cancer Genome Atlas (TCGA) AML samples (n=194) run on Illumina 450k arrays. We used RNA-seq data to study gene expression changes associated with hypomethylator phenotypes (HP). Analysis was done using R. Results: Genome-wide analysis of CpG sites that are highly methylated in normal blood, and variably methylated in AML (β-value standard deviation in AML > 0.2; average β-value in normal blood > 0.8) revealed two distinct HPs by hierarchical clustering: Good-risk (GR) HP which included favorable cytogenetics, and DNMT-HP, which was enriched for DNMT3A mutations. We refined DNA methylation signatures of each HP cluster by differential methylation analysis and re-classified patients accordingly. Strikingly, all patients with t(8;21), inv(16), or t(15;17) belonged to the GR-HP+ group, suggesting that a common epigenetic thread connects these otherwise disparate genetic aberrations. From a clinical perspective GR-HP+ patients were younger than GR-HP- patients, and had significantly longer overall survival (median OS, years: GR-HP+ = Not reached; GR-HP- = 1.00; P < 0.001). In contrast, DNMT-HP+ cases were statistically equivalent to DNMT-HP- except for an enrichment for higher WBC counts, including no difference in survival (median OS, years: DNMT-HP+ = 0.92; DNMT-HP- = 1.34; P = 0.27). From an epigenetic perspective the two HP clusters harbored distinct DNA methylation changes; although both favored hypomethylation within non-CpG islands relative to CpG islands, the enrichment was more pronounced for DNMT-HP (Odds ratio: hypomethylated CpG islands/hypomethylated non-CpG islands, GR-HP = 0.64; DNMT-HP = 0.18). Genetic analysis revealed that GR-HP+ leukemia had wild-type IDH, DNMT3A, and NPM1 genes. In contrast, DNMT-HP+ AML had significantly more FLT3, NPM1, and DNMT3A mutations compared to DNMT-HP- patients. RNA-seq revealed significant up-regulation of genes in both HP phenotypes (216, and 150 genes for GR-HP and DNMT-HP, respectively at FDR < 0.01 and FC > 2). Pathway analysis of these genes revealed enrichments for ion channels and the complement pathway in DNMT-HP, and for nervous system and developmental genes in GR-HP. Conclusions: Our data suggest that two HPs exist in AML with unique epigenetic and transcriptomic signatures. The striking association between GR-HP and different favorable cytogenetic changes suggests that a common set of epigenetic features may contribute to improved survival in these patients. Citation Format: Andrew D. Kelly, Jozef Madzo, Priyanka Madireddi, Patricia Kropf, Charly R. Good, Jaroslav Jelinek, Jean-Pierre J. Issa. A DNMT3A-independent hypomethylator phenotype is a unifying epigenetic signature of AML with good risk cytogenetics [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5382. doi:10.1158/1538-7445.AM2017-5382
- Published
- 2017
- Full Text
- View/download PDF
35. Abstract 4677: Integrated molecular profiles and guadecitabine response in relapsed/refractory acute myeloid leukemia
- Author
-
Jean Pierre J. Issa, Hagop M. Kantarjian, Patricia Kropf, Woonbok Chung, Pietro Taverna, Jaroslav Jelinek, Sue Naim, Mohammad Azab, and Andrew Kelly
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,CpG Island Methylator Phenotype ,Myeloid leukemia ,Cancer ,Methylation ,Biology ,medicine.disease ,medicine.disease_cause ,Leukemia ,CpG site ,Internal medicine ,DNA methylation ,medicine ,KRAS - Abstract
Epigenetic patterns of DNA methylation are frequently altered in acute myeloid leukemia (AML). Guadecitabine is a novel next generation hypomethylating drug with a demonstrated clinical activity in elderly AML. In our previous study of guadecitabine, we identified in relapsed/refractory (r/r) AML patients a gene expression signature (high DNMT3B, low P15, and low CDA) associated with reduced LINE-1 demethylation and resistance to the drug. Since pharmacodynamics only explains a small portion of the variability in response to guadecitabine, we suggest that intrinsic genetic and epigenetic characteristics can distinguish responders from non-responders. We analyzed genome-wide methylation patterns and screened 54 genes frequently mutated in leukemia in DNA from pre-treatment blood or bone marrow from 119 patients with r/r AML enrolled in guadecitabine phase I/II trials. Blood samples from 49 healthy donors were used as controls. We also examined expression of a panel of genes (CDA, P15, P21, DNMT3B, DNMT3A, DNMT1, and CTCF) at baseline by quantitative RT-PCR. Global DNA methylation at pre/post treatment was estimated by bisulfite-pyrosequencing of the LINE-1 repetitive sequence. For comparing methylation profiles at CpG islands (CGI), we performed hierarchical clustering analysis of differentially methylated 2794 CGI sites in 119 r/r AML and 49 healthy controls. Hierarchical clustering split them into 3 main groups of normal-like cluster (n=36), intermediate (n=41) and CpG island methylator phenotype (CIMP) like cluster (n=42). Normal-like cluster had a higher response rate (39 %) compared to intermediate (17%) and CIMP- like cluster (15%) (p=0.025, Fisher’s exact test). Demethylation rate of LINE-1 was lower in CIMP-like cluster than in normal-like and intermediate cluster (average demethylation -19 ± 2 % in CIMP-like cluster vs. -31 ± 2 % in normal-like vs. -23 ± 2 % in intermediate cluster, p=0.0007, One-way ANOVA). Mutation screening revealed frequent alterations affecting signaling pathways (CSF3R, KIT, KRAS, NRAS and FLT3) in the CIMP-like cluster (62%) compared to normal-like (11%) and intermediate cluster (13%) (p Citation Format: Woonbok Chung, Andrew D. Kelly, Patricia Kropf, Pietro Taverna, Sue Naim, Mohammad Azab, Jaroslav Jelinek, Hagop M. Kantarjian, Jean-Pierre J. Issa. Integrated molecular profiles and guadecitabine response in relapsed/refractory acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4677. doi:10.1158/1538-7445.AM2017-4677
- Published
- 2017
- Full Text
- View/download PDF
36. (P088) Splenic Irradiation for Splenomegaly: A Meta-Analysis
- Author
-
Shelly B. Hayes, Patricia Kropf, Nicholas G. Zaorsky, Stefan K. Barta, Joshua E. Meyer, Graeme R. Williams, and Nestor F. Esnaola
- Subjects
Cancer Research ,medicine.medical_specialty ,Radiation ,Oncology ,business.industry ,Internal medicine ,Meta-analysis ,medicine ,Radiology, Nuclear Medicine and imaging ,Splenic irradiation ,business ,Gastroenterology - Published
- 2017
- Full Text
- View/download PDF
37. Genetic Determinants of Response to Guadecitabine (SGI-110) in AML
- Author
-
Gail J. Roboz, Andrew Kelly, Pietro Taverna, Woonbok Chung, Patricia Kropf, Jean Pierre J. Issa, Hagop M. Kantarjian, Sue Naim, Karen W.L. Yee, Mohammad Azab, and Jaroslav Jelinek
- Subjects
Oncology ,Neuroblastoma RAS viral oncogene homolog ,medicine.medical_specialty ,Mutation rate ,education.field_of_study ,Mutation ,Myeloid ,Immunology ,Population ,Decitabine ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,Bioinformatics ,Biochemistry ,medicine.anatomical_structure ,Internal medicine ,Pharmacodynamics ,medicine ,KRAS ,education ,medicine.drug - Abstract
Background: Guadecitabine is a second generation hypomethylating drug with improved pharmacokinetics and pharmacodynamics compared to decitabine and demonstrated clinical activity in both treatment naïve (tn) and relapsed-refractory (rr) AML. Previous studies reported similar response rates to guadecitabine in different cytogenetic subsets but it remains unknown whether this extends to genetic changes. Methods: Pre-treatment blood or bone marrow derived DNA was available for analysis from a total of 220 patients with AML enrolled on guadecitabine phase I-II trials (121 rrAML and 99 tnAML). We included only patients treated at therapeutic doses. Mutations in a panel of 54 genes was studied by the TruSight Myeloid Sequencing Panel (Illumina) and deep sequencingon the Illumina HiSeq platform. FLT3 mutations were separately determined by PCR analysis. The sequence data was analyzed for mutations using the TruSeq Amplicon Application in the Illumina BaseSpace Suite. Putative mutation calls were further filtered by sequencing read quality, minimum variant allelic fraction, and presence in the dbSNP and COSMIC databases. Results: In aggregate, responses to guadecitabine in rrAML were 15 CR (12%), 12 PR/CRi/CRp and 94 non responders (NR) and in tnAML, responses were 34 CR (34%), 21 PR/CRi/CRp and 44 NR. Overall, a median of 1 (range 0 - 5) mutation was present in each patient, with no significant differences between tn and rr AML. The most frequently mutated genes were ASXL1 (16.8%), TET2 (14.1%), IDH2 (10.9%), NPM1 (10%), RUNX1 (9.5%), DNMT3A (9.1%), NRAS (9.1%), FLT3-ITD (8.6%), U2AF1 (8.2%), IDH1 (6.8%), TP53 (5%), and KRAS (4.5%). The distribution of mutations was as expected for a group of patients with rrAML and elderly tnAML. We used Fisher's exact tests to compare mutation frequencies between patients who achieved CR and those who did not achieve CR. When we evaluated rrAML and tnAML separately, none of the genes showed a significantly different mutation rate between response subgroups. We then examined the population as a whole (N=220) and found that mutations in NRAS were significantly lower in patients who achieved CR (0/49) compared to those who did not (20/171, p=0.009). NRAS and KRAS mutations were inversely correlated, and when we considered the two genes together, mutations were present in 1/49 CR patients compared to 28/171 non-CR patients (p=0.007). Overall, CR rate was 3.4% in patients with RAS mutations compared to 25.1% in patients without such mutations. There was a similar significant trend for IDH2 mutations to be lower in CR patients (1/48) compared to non-CR patients (23/172, p=0.02) but this was not seen for IDH1. None of the mutations in other epigenetic regulators (DNMT3A, ASXL1, EZH2, TET2, U2AF1 or WT1) were significantly different between CR and non-CR patients individually or when we considered mutations in any of 8 epigenetic regulators (mutated in 22/49 CR patients vs. 93/171 non-CR patients, p=0.26). RAS mutations were higher in rrAML (22/121, 18.2%) than in tnAML (7/99, 7.1%, p=0.017) which may explain the lower CR rate in this group. Patients with PR/CRi/CRp were genetically similar to NR. Conclusions: In patients with AML treated with guadecitabine, RAS pathway mutations and IDH2 mutations are associated with a lower likelihood of achieving a CR. Disclosures Kropf: Celgene: Consultancy; Takeda: Consultancy. Roboz:Cellectis: Research Funding; Agios, Amgen, Amphivena, Astex, AstraZeneca, Boehringer Ingelheim, Celator, Celgene, Genoptix, Janssen, Juno, MEI Pharma, MedImmune, Novartis, Onconova, Pfizer, Roche/Genentech, Sunesis, Teva: Consultancy. Yee:Novartis Canada: Membership on an entity's Board of Directors or advisory committees, Research Funding. Taverna:Astex Pharmaceuticals: Employment. Naim:Astex Pharmaceuticals: Employment. Azab:Astex Pharmaceuticals, Inc.: Employment. Issa:Teva Pharmaceutical Industries: Consultancy; Astex Pharmaceuticals: Consultancy.
- Published
- 2016
- Full Text
- View/download PDF
38. Improved Survival for MDS/CMML Patients Treated with the Combination of Decitabine (DAC) and Arsenic Trioxide (ATO) in a Phase II Adaptive Three Arm Randomization Study: DAC Alone Vs. DAC +/- Carboplatin or ATO
- Author
-
Patricia Kropf, Hagop M. Kantarjian, Henry C. Fung, Philip A. Pancari, Xuelin Huang, Raji Shameem, Woonbok Chung, Jean Pierre J. Issa, and Lianchun Xiao
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Myelodysplastic syndromes ,Immunology ,Decitabine ,Phases of clinical research ,Chronic myelomonocytic leukemia ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Carboplatin ,chemistry.chemical_compound ,Hypomethylating agent ,Tolerability ,chemistry ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Introduction: DAC is a hypomethylating agent FDA approved for the treatment of advanced myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML).Despite its clinical activity, the duration of response is limited and prognosis at relapse is poor. In pre-clinical studies, we identified potent epigenetic effects of ATO alone and in combination with DAC, and also found that carboplatin (Carbo) can enhance gene reactivation in combination with DAC. We therefore initiated a randomized phase 2 clinical trial (NCT02188706) to compare the safety and efficacy of DAC in combination with either Carbo or ATO as compared to single agent DAC in patients with MDS/CMML and Acute Myeloid Leukemia. Here we present updated results for the MDS/CMML cohort. Methods: Patients with MDS/CMML INT-1 were randomized to one of three regimens: DAC 20 mg/m2 days 1-5, (DAC), DAC as above and Carbo AUC 5 on day 8 (DAC/Carbo), or DAC as above and ATO 0.15 mg/kg days 1-5 (DAC/ATO). We used adaptive randomization based on response rate which started after 10 patients were equally randomized to each arm. Cycles were scheduled every 28 days for a minimum of 4 cycles. Dose reductions/delays were allowed based on response and tolerability. Patients remained on treatment as long as they continued to benefit without toxicity. The primary endpoint was the composite response rate: the complete response (CR: complete response, mCR: marrow complete response and CRi: complete response with incomplete blood count recovery) and partial response (PR) rates using the modified IWG 2006 criteria. Secondary endpoints included median overall survival (OS) and safety. DNA methylation changes were measured using LINE1 bisulfite/pyrosequencing. Results: As of July 2016, 42 patients (MDS = 40, CMML=2) have been enrolled on study. Median age was 71 years (range, 35 to 84 years). There was no statistically significant difference in patients' characteristics between the three arms. Median number of cycles for the treatment arms was 4 (range, 1-15). Response data was evaluable for 36 patients (8 on DAC alone, 5 on DAC/Carbo and 23 on DAC/ATO). Composite responses were seen in 3/8 patients on DAC (37.5%), 0/5 patients on DAC/Carbo (0%, p=0.23 compared to DAC alone) and 12/23 patients on DAC/ATO (52.2%, p=0.69 compared to DAC alone). Median OS among MDS/CMML patients receiving DAC/ATO (17.8 months) was improved compared to the DAC/Carbo (3.9 months) and DAC (9.8 months) arms (p=0.01) [Figure 1]. There was not a significant difference in high-grade (≥ grade 3) toxicity between the three arms, with grade 3 neutropenia and thrombocytopenia as the most common side effects. DNA methylation analysis showed equivalent demethylation in all the arms, suggesting that the epigenetic effects of ATO were independent of DNA methylation, which was consistent with pre-clinical studies. Conclusion: Our results demonstrate higher response rates and a significant survival benefit with the combination of DAC and ATO in comparison to DAC alone in patients with MDS/CMML. Disclosures Kropf: Celgene: Consultancy; Takeda: Consultancy. Fung:Amgen: Consultancy; Genzyme: Consultancy. Kantarjian:ARIAD: Research Funding; Bristol-Myers Squibb: Research Funding; Amgen: Research Funding; Pfizer Inc: Research Funding; Delta-Fly Pharma: Research Funding; Novartis: Research Funding. Issa:Astex Pharmaceuticals: Consultancy; Teva Pharmaceutical Industries: Consultancy.
- Published
- 2016
- Full Text
- View/download PDF
39. Clinicopathologic features and survival outcomes of secondary diffuse large b-cell lymphoma after a primary solid malignancy: SEER propensity-matched analysis
- Author
-
Muhammad Saad Hamid, Patricia Kropf, Chethan Ramamurthy, Pooja Ghatalia, Raji Shameem, and Philip A. Pancari
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Prior diagnosis ,medicine.disease ,Malignancy ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,Propensity score matching ,medicine ,In patient ,business ,neoplasms ,Diffuse large B-cell lymphoma - Abstract
e19022Background: Outcomes of secondary DLBCL (sDLBCL) in patients with a prior diagnosis of a solid malignancy compared to de novo DLBCL (dnDLBCL) are not well understood. We sought to determine f...
- Published
- 2016
- Full Text
- View/download PDF
40. Comparing Cyclophosphamide with G-CSF and Plerixafor with G-CSF As Stem Cell Mobilization (SCM) Regimens for Multiple Myeloma (MM) Patients: An Evaluation of Mobilization Efficacy and Toxicity
- Author
-
Philip A. Pancari, Henry C. Fung, Stefan K. Barta, Mary Ellen Martin, Carlyn Rose C. Tan, John Ulicny, and Patricia Kropf
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,Mobilization ,Cyclophosphamide ,Stem cell mobilization ,business.industry ,Plerixafor ,Hematology ,medicine.disease ,Internal medicine ,Toxicity ,medicine ,business ,Multiple myeloma ,medicine.drug - Published
- 2016
- Full Text
- View/download PDF
41. Long-term results of chemoimmunotherapy with low-dose fludarabine, cyclophosphamide and high-dose rituximab as initial treatment for patients with chronic lymphocytic leukemia
- Author
-
Ahmad A. Tarhini, Mathew Sulecki, Suzanne Lentzsch, L. Pietragallo, Diana Lenzner, Patricia Kropf, Stanley M. Marks, Dhaval R. Mehta, Kenneth A. Foon, and Michael Boyiadzis
- Subjects
Oncology ,Adult ,medicine.medical_specialty ,Time Factors ,Cyclophosphamide ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,Immunology ,Neutropenia ,Biochemistry ,Maintenance Chemotherapy ,Antibodies, Monoclonal, Murine-Derived ,Clinical Trials, Phase II as Topic ,immune system diseases ,Chemoimmunotherapy ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Combined Modality Therapy ,Humans ,Neoadjuvant therapy ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Analysis ,Neoadjuvant Therapy ,Fludarabine ,Rituximab ,Immunotherapy ,business ,Vidarabine ,medicine.drug ,Follow-Up Studies - Abstract
To the editor: Chemoimmunotherapy with fludarabine (F), cyclophosphamide (C), and rituximab (R) is currently considered the gold standard first-line therapy for chronic lymphocytic leukemia (CLL).[1][1] In an attempt to reduce the neutropenia and maintain the high response rate of standard-dose FCR
- Published
- 2012
42. Spontaneous graft versus host disease occurring in a patient with multiple myeloma after autologous stem cell transplant
- Author
-
Patricia Kropf, Robert Emmons, MaryEllen Martin, Manish Sharma, Mangan Kf, Thomas R. Klumpp, Christian Joseph Fidler, and Meina Lu
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,Gastroenterology ,Transplantation, Autologous ,Fatal Outcome ,Internal medicine ,medicine ,Humans ,Multiple myeloma ,Skin ,business.industry ,Hematopoietic Stem Cell Transplantation ,Immunosuppression ,Hematology ,Middle Aged ,medicine.disease ,Rash ,Transplantation ,Gastrointestinal Tract ,surgical procedures, operative ,Graft-versus-host disease ,Vomiting ,Female ,medicine.symptom ,Complication ,business ,Multiple Myeloma - Abstract
Graft versus host disease (GVHD) is a common complication of allogeneic transplant. Acute GVHD primarily affects the skin, liver, and GI tract generally within the first 100 days after transplant. GVHD following an allogeneic transplant occurs as a result of donor T-cell recognition of host alloantigens. In contrast, patients undergoing ASCT are not subjected to the genetic disparity that occurs with allogeneic transplant, and in principal, should not develop this proinflammatory response. A clinical syndrome, however, has been described in patients following autologous transplant that shares the same features as GVHD occurring in recipients post-allogeneic transplant [1-3]. Previously reported cases have described skin, liver, and GI tract manifestations consistent with what is seen in allogeneic GVHD. Biopsies of the skin and GI tract mucosa have demonstrated similar histological features as well. Interestingly, the majority of reported cases seem to occur in patients with multiple myeloma undergoing consolidative ASCT. Historically, however, these patients have been described as having a relatively benign course with mild skin rash, nausea, vomiting, and/or diarrhea that is responsive to immunosuppression. In this article, we present a case of fatal, spontaneous GVHD in a patient with multiple myeloma following ASCT.
- Published
- 2011
43. Src family kinase gene targets during myeloid differentiation: identification of the EGR-1 gene as a direct target
- Author
-
Daniel Johnson, Patricia Kropf, R Duan, J E Jones, and L Wang
- Subjects
Cancer Research ,Myeloid ,Cellular differentiation ,Antineoplastic Agents ,Tretinoin ,Biology ,Article ,Leukemia, Promyelocytic, Acute ,medicine ,Tumor Cells, Cultured ,Humans ,Src family kinase ,RNA, Messenger ,Gene ,Early Growth Response Protein 1 ,Oligonucleotide Array Sequence Analysis ,Genetics ,Gene targets ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Cell Differentiation ,Hematology ,Cell biology ,body regions ,Gene expression profiling ,medicine.anatomical_structure ,Pyrimidines ,src-Family Kinases ,Oncology ,Identification (biology) ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Src family kinase gene targets during myeloid differentiation: identification of the EGR-1 gene as a direct target
- Published
- 2009
44. Multivariate Analysis Identifies Significant Correlations Between Baseline Biomarkers, DNA Demethylation and Clinical Responses in 122 Patients Treated in a Phase 1/2, Study of Guadecitabine (SGI-110), in the Treatment of Relapsed/Refractory Acute Myeloid Leukemia (r/r AML)
- Author
-
Mohammad Azab, Gail J. Roboz, Shashank Rohatagi, Pietro Taverna, Hagop M. Kantarjian, Patricia Kropf, Woonbok Chung, Srikanth Gottipati, and Jean Pierre J. Issa
- Subjects
Oncology ,NPM1 ,medicine.medical_specialty ,biology ,business.industry ,Immunology ,C-reactive protein ,Decitabine ,Phases of clinical research ,Cell Biology ,Hematology ,Bioinformatics ,Biochemistry ,Regimen ,Hypomethylating agent ,Pharmacodynamics ,Internal medicine ,DNA methylation ,medicine ,biology.protein ,business ,medicine.drug - Abstract
Introduction: Guadecitabine (SGI-110) is a novel subcutaneous (SC) next generation hypomethylating agent (HMA) designed as a dinucleotide of decitabine (DAC) and deoxyguanosine that is resistant to degradation by cytidine deaminase (CDA) and results in prolonged in vivo exposure to its active moiety DAC. The differentiated pharmacokinetic profile offers the potential of improved biological and clinical activity and safety over currently available HMAs. In the Phase 1 study, patients with r/r AML were treated at escalating doses of guadecitabine. In the phase 2 study, r/r AML patients were randomized to receive guadecitabine at 60 mg/m2 or 90 mg/m2 SC daily for 5 days (dailyx5). In a separate cohort, patients were treated with 60 mg/m2 SC daily for 10 days (dailyx10: days 1-5 and 8-12) for up to 4 cycles followed by subsequent cycles of the dailyx5 regimen. All regimens were dosed with a 28 day treatment cycle. We have reported the clinical efficacy and safety results from the Phase 1 dose-escalation study in AML and MDS (Issa et al, Lancet Oncology 2015) and the Phase 2 randomized dose-response study in r/r AML at 2 doses (60 and 90 mg/m2) in a 5-day regimen (Kantarjian et al, ASH 2013) and 60 mg/m2 in the 10-day regimen (Griffiths et al, ASCO 2014). Here, we report the associations between clinical responses and global DNA demethylation assessed by LINE1 assay, baseline expression of a panel of 7 genes (CDA, P15, P21, DNMT3B, DNMT3A, DNMT1 and CTCF) assessed by qRT-PCR, and FLT3-ITD/NMP1 mutations. Methods: Samples from 122 patients with r/r AML were analyzed (27 from phase 1 study treated at 36 m/m2/d or higher, 47 from the dailyx5 and 48 from the dailyx10 regimen of the phase 2). Global DNA methylation at baseline and after treatment, were estimated using bisulphite-pyrosequencing on the LINE-1 repetitive sequence for assessing guadecitabine pharmacodynamic (PD) effects. LINE-1 methylation time-course profiles were available in 117 out of 122 analyzed patients. Multivariate analysis was performed using nonlinear ensemble decision tree-based classification algorithms (Random Forests and Gradient Boosting Trees); the algorithms were applied to rank demographic features, treatment schedule features, baseline expression of 7 genes, FLT3-ITD and NPM1 mutations, LINE-1 methylation profiles and baseline hematological features as predictors of clinical response and to measure their directionality. All reported percentages are a measure of the average relative contribution of each feature in predicting response across boosted trees and are normalized across features. Results: In the 122 patients analyzed, the median age was 59.6 (range, 23-86), 75 were males (61.5%). Overall, peak LINE-1 demethylation generally occurred on day 8 after daily x 5 treatment or on day 8 or 15 after daily x 10 treatment. Overall, the maximum peak LINE-1 demethylation was -23.8 % ± 1.24. In 122 r/r AML patients, 28 showed Complete Response (23%, 14 CR and 14 CRi/CRp). High baseline CDA gene expression in peripheral blood was consistently ranked the main predictor of clinical response (mean for responders~1.76 and non-responders~0.12) irrespective of patient gender, dose and dosing cycles, and contributed for 22% of the model accuracy in predicting responses to guadecitabine. Expression of the genes CTCF (11%), DNMT3B (7%) and P21 (6%) were other genes that contributed > 5% in predicting clinical response. Age (8%) was also a significant predictor of response. FLT3-ITD mutations and NPM1 mutations were not significant predictors of response. In an integrated analysis, where methylation levels and gene expression were ranked together on their power to predict clinical response, LINE-1 methylation percent change from baseline on days 8(6%) and 22(19%), age (16%), CTCF (9%), CDA(7%), P21(6.5%) gene expression at baseline had greater than 5% contribution in predicting clinical response. A similar analysis was then performed to rank features on their power to predict day 8 methylation; CDA (28%), DNMT1 (8.5%), P15 (7.5%), and CTCF (7%) gene expression at baseline and age (7%) had greater than 5% predictive power. Conclusions: In conclusion, in r/r AML patients, global DNA demethylation was strongly associated with clinical responses to guadecitabine and analysis of baseline gene expression can identify trends that might enrich for r/r AML patients more likely to respond to HMA therapy with guadecitabine. Disclosures Gottipati: Otsuka Pharmaceutical Development & Commercialization, Inc.: Employment. Rohatagi:Otsuka Pharmaceutical Development & Commercialization, Inc.: Employment. Chung:Astex Pharmaceuticals, Inc.: Research Funding. Taverna:Astex Pharmaceuticals, Inc.: Employment. Kropf:Teva Pharmaceuticals: Consultancy. Azab:Astex Pharmaceuticals, Inc.: Employment. Issa:Astex Pharmaceuticals, Inc.: Consultancy; Janssen: Consultancy.
- Published
- 2015
- Full Text
- View/download PDF
45. Comparison of Efficacy and Safety Results in 103 Treatment-Naïve Acute Myeloid Leukemia (TN-AML) Patients Not Candidates for Intensive Chemotherapy Using 5-Day and 10-Day Regimens of Guadecitabine (SGI-110), a Novel Hypomethylating Agent (HMA)
- Author
-
James N. Lowder, Raoul Tibes, Laksmi Wilson, Wendy Stock, Jean Pierre J. Issa, Pietro Taverna, David A. Rizzieri, Patricia Kropf, Karen W.L. Yee, Jesus G. Berdeja, Elias Jabbour, Katherine Walsh, Guillermo Garcia-Manero, Mohammad Azab, Hagop M. Kantarjian, Nikolai A. Podeltsev, Elizabeth A. Griffiths, Casey O'Connell, Gail J. Roboz, Yong Hao, Michael R. Savona, and Todd L. Rosenblat
- Subjects
medicine.medical_specialty ,business.industry ,Surrogate endpoint ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Regimen ,Tolerability ,Median follow-up ,Internal medicine ,Cohort ,medicine ,Clinical endpoint ,business ,Febrile neutropenia - Abstract
Introduction: Guadecitabine (SGI-110) is a novel next-generation HMA administered as a small volume subcutaneous (SC) injection which results in extended decitabine exposure. Phase 2 studies have been conducted in TN-AML patients who were not candidates for intensive chemotherapy using two different doses and schedules of guadecitabine. We report here a comparative efficacy and safety analysis of the 5-day and 10-day regimens. Methods: TN-AML patients who were not candidates for intensive chemotherapy based on age (≥ 65 y), poor performance status (PS 2), comorbidities, or poor risk cytogenetics were enrolled in 2 separate treatment cohorts in the Phase 2 study. In the first cohort, patients were randomized (1:1) to either 60 mg/m2/d or 90 mg/m2/d on Days 1-5 (5-day regimen). In the second cohort, patients were treated with 60 mg/m2/d on Days 1-5 and Days 8-12 (10-day regimen) for up to 4 cycles, followed by 60 mg/m2/d Days 1-5 in subsequent cycles. Cycles were scheduled every 28 days for both regimes with dose reductions/delays allowed based on response and tolerability. Patients remained on treatment as long as they continued to benefit with no unacceptable toxicity. The primary endpoint was the composite Complete Response (CRc): Complete Response (CR) + CR with incomplete platelet recovery (CRp) + CR with incomplete neutrophil recovery (CRi) using modified International Working Group (IWG) criteria (Cheson et al, 2003). Secondary endpoints included overall survival (OS), and safety. Results: There was no difference in efficacy or safety between 60 and 90 mg/m2/d on the 5-day regimen (Yee et al, European Hematology Association meeting 2014, S647), so data are reported here for the two doses combined on the 5-day cohort. There were 51 patients treated in the 5-day regimen cohort and 52 treated with the 10-day regimen. There was no statistically significant difference in patient characteristics between the 2 regimens; median age 77.9 vs. 77.3 years ; male 59% vs. 65%; PS 2 or higher 35% vs. 40%; median BM blasts 40.0% vs. 49.5%; poor risk cytogenetics 46% vs. 43% for the 5-day and 10-day cohorts, respectively. Follow up of the 10-day cohort patients was shorter as it started after completion of enrolment of the 5-day cohort. The median follow up was 25.7 and 12.4 months and median number of cycles was 5 (range 1-26) and 3 (range 1-13) for the 5-day and 10-day cohorts, respectively. There was no significant difference in the primary efficacy endpoint, CRc, between the 2 regimens (p=0.43). CRc was achieved in 29/51 patients (57%) on the 5-day regimen (19 CR, 3 CRp, and 7 CRi) and in 25/52 patients (48%) on the 10-day regimen (16 CR, 5 CRp, and 4 CRi). Median OS was 10.5 and 8.7 months for the 5-day and 10-day cohorts, respectively (p=0.89). The 30, 60, and 90-day all-cause mortality rates were not statistically significant between the two cohorts: 5.9%, 15.7%, and 21.6% on the 5-day regimen and 1.9%, 17.3%, and 28.8% on the 10-day regimen. The most common Grade ≥3 AEs regardless of relationship to guadecitabine were: febrile neutropenia 59% vs. 60%, thrombocytopenia 47% vs. 38%, neutropenia 39% vs. 33%, anemia 27% vs. 19%, pneumonia 24% vs. 27%; and sepsis 12% vs. 19%, for the 5-day and 10-day cohorts respectively, none of which was statistically significant. Fifteen patients remain on treatment (5 from the 5-day cohort and 10 from the 10-day cohort). Conclusions: Guadecitabine is clinically active with a good safety profile in TN-AML patients not candidates for intensive chemotherapy. Unlike in relapsed/refractory AML, where the 10-day regimen of guadecitabine showed a trend toward improved efficacy (Roboz et al, Annals of Oncology 25 Supplement 4, 2014), there was no significant difference in either efficacy or safety between the 5-day and 10-day regimens in newly diagnosed AML patients. Guadecitabine 60 mg/m2/d SC Days 1-5 is currently being investigated in an 800-patient multicenter randomized phase 3 study in TN-AML patients unfit to receive intensive chemotherapy (ASTRAL-1 Phase 3 clinical trial: ClinicalTrials.gov reference NCT02348489). Disclosures Kropf: Teva Pharmaceuticals: Consultancy. O'Connell:Celgene: Speakers Bureau; Incyte: Membership on an entity's Board of Directors or advisory committees. Griffiths:Astex: Research Funding; Alexion Pharmaceuticals: Honoraria; Celgene: Honoraria. Rizzieri:Teva: Other: ad board, Speakers Bureau; Celgene: Other: ad board, Speakers Bureau. Stock:Gilead: Membership on an entity's Board of Directors or advisory committees. Savona:Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees. Rosenblat:Astex Pharmaceuticals, Inc.: Research Funding. Berdeja:Celgene: Research Funding; Onyx: Research Funding; BMS: Research Funding; Abbvie: Research Funding; Array: Research Funding; Curis: Research Funding; Acetylon: Research Funding; MEI: Research Funding; Janssen: Research Funding; Novartis: Research Funding; Takeda: Research Funding. Wilson:Astex Pharmaceuticals, Inc.: Employment. Lowder:Astex Pharmaceuticals, Inc.: Employment. Taverna:Astex Pharmaceuticals, Inc.: Employment. Hao:Astex Pharmaceuticals, Inc.: Employment. Azab:Astex Pharmaceuticals, Inc.: Employment. Issa:Janssen: Consultancy; Astex Pharmaceuticals, Inc.: Consultancy.
- Published
- 2015
- Full Text
- View/download PDF
46. Clinicopathologic Features and Survival Outcomes of Secondary Mantle Cell Lymphoma after Primary Solid Malignancy
- Author
-
Nasheed Hossain, Muhammad Saad Hamid, Patricia Kropf, Raji Shameem, and Jeffrey Mufson
- Subjects
medicine.medical_specialty ,Proportional hazards model ,business.industry ,Endometrial cancer ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Malignancy ,Biochemistry ,Gastroenterology ,Cancer registry ,Internal medicine ,medicine ,Surveillance, Epidemiology, and End Results ,Pacific islanders ,Mantle cell lymphoma ,Stage (cooking) ,business - Abstract
Background: Mantle Cell Lymphoma (MCL) is a rare sub-type of Non-Hodgkin's Lymphoma (NHL). Outcomes of secondary MCL (sMCL) in patients with a prior diagnosis of a solid malignancy as compared to de novo MCL are not well understood. Differences may be attributable to age, gender, race/ethnicity, extra-nodal involvement, and stage at initial diagnosis. We sought to determine differences in outcome by analyzing data from a large nationwide cancer registry. Methods: The Surveillance Epidemiology and End Results (SEER) database (1973-2012) was used to detect MCL (ICD-O-3 code: 9673/3) adult (>18 years) cases. The variable "First Malignant Primary Indicator" was used to differentiate between de novo and sMCL cases. Primary solid malignancy subtypes categorized included prostate, breast, colorectal, lung bladder, renal, head and neck, thyroid, testicular, and endometrial cancer. Selected sMCL cases included patients with a diagnosis of a primary solid malignancy with a latency period ≥ 2 months. Overall survival (OS) was calculated using the cox regression model to determine the impact of sMCL on survival, adjusting for age at diagnosis, race/ethnicity, primary solid malignancy subtype type and latency period. Chi square test was utilized to ascertain for any significant difference between de novo MCL and sMCL cases. Results: Overall, 8,889 patients were included: 875 (9.8%) sMCL and 8,014 (90.2%) de novo MCL cases. For de novo MCL, 67.1% were males. Race/ethnicity in descending frequency consisted of Non-Hispanic Whites (NHW) (81.8%), Hispanics (8.4%), Blacks (4.4%), Asian and Pacific Islanders(API)(4.1%) and American Indian/Native Indian (AI) (0.4%). The most frequent stage at initial diagnosis for de novo MCL was stage IV (57.9%) followed by stage III (13.4%), stage II (8.1%) and stage I (10.0%). Males consisted of 75.4% of sMCL cases. Cases of sMCL categorized by race/ethnicity in descending frequency were NHW (87.3%), Hispanics (5.1%), Blacks (4.2%), API (3.2%) and AI (0.1%). Stage at diagnosis for sMCL was most frequently stage IV (54.6%), followed by stage III (15.8%), stage II (10.9%) and stage I (11.7%). Compared to de novo MCL, sMCL cases were more likely to occur in patient's ≥75 years (48.3% vs. 28.7%, p Conclusion: Our analysis demonstrates that elderly age appears to be significantly more common in sMCL cases as compared to de novo MCL. By contrast, race/ethnicity, and extra-nodal involvement were similar between the two groups. Survival was worse in sMCL compared to de novo MCL. This may be due to differences in treatment preferences. Further studies are necessary to elucidate the etiology for such differences in outcomes. Disclosures Kropf: Teva Pharmaceuticals: Consultancy.
- Published
- 2015
- Full Text
- View/download PDF
47. Superior Response Rates for Patients with Myeloid Malignancies Treated with the Combination of Decitabine (DAC) and Arsenic Trioxide (ATO) in a Phase II Adaptive Three Arm Randomization Study: DAC +/- Carboplatin or ATO
- Author
-
Patricia Kropf, Huang Xuelin, Henry C. Fung, Philip A. Pancari, Hagop M. Kantarjian, Lianchun Xiao, and Jean Pierre J. Issa
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,Myelodysplastic syndromes ,medicine.medical_treatment ,Immunology ,Decitabine ,Phases of clinical research ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Carboplatin ,Surgery ,chemistry.chemical_compound ,Hypomethylating agent ,Tolerability ,chemistry ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
Background: Epigenetic therapy is the mainstay of treatment for patients with advanced myelodysplastic syndromes (MDS) and for patients with acute myeloid leukemia (AML) who are not candidates for intense chemotherapy. Decitabine (DAC) is an FDA approved hypomethylating agent for the treatment of MDS and has clinical activity in AML. Despite DAC's activity in MDS and AML, the duration of response is limited, and once patients relapse the prognosis is poor. In pre-clinical studies, we identified potent epigenetic effects of arsenic trioxide (ATO) alone and in combination with DAC, and also found that carboplatin (Carbo) can enhance gene reactivation by DAC. We therefore initiated a randomized phase 2 clinical trial (NCT02188706) to compare the safety and efficacy of DAC in combination with either Carbo or ATO as compared to single agent DAC in patients with MDS and AML. Methods: Patients with MDS INT-1 or above or patients with AML that was relapsed or refractory or considered unfit for intense chemotherapy based on age (>60 years), poor performance status (PS2) or comorbidities were randomized to one of three regimens: DAC 20 mg/m2 days 1-5, DAC as above and Carbo AUC 5 on day 8, or DAC as above and ATO 0.15 mg/kg days 1-5. We used adaptive randomization based on response rate, and the adaptive randomization started after 10 patients were randomized to each arm equally. Cycles were scheduled every 28 days for a minimum of 4 cycles. Dose reductions/delays were allowed based on response and tolerability. Patients remained on treatment as long as they continued to benefit without toxicity. The primary endpoint was the composite response rate: the complete and partial response rates using the modified IWG 2006 criteria in MDS and IWG 2003 criteria in AML. Secondary endpoints included survival (median, one year), safety and an evaluation of the epigenetic effects of each arm. Results: 42 patients have been enrolled on study as of August 2015 (22 AML and 20 MDS). Median age was 65 years (range, 30 to 85 years). There was no statistically significant difference in patients' characteristics between the three arms. Twenty-three (60%) of the patients were previously treated and had relapsed/refractory disease. 38 patients were evaluable for response (13 on DAC alone, 11 on DAC/Carbo and 14 on DAC/ATO). There was not a significant difference in toxicity between the arms, with Grade 3 neutropenia and Grade 3 thrombocytopenia as the most common side effects. Responses (CR, Cri, PR) were seen in 3/13 patients on DAC alone (23%), 5/11 patients on DAC/Carbo (45%, p=0.18 compared to DAC alone) and 10/14 patients on DAC/ATO (71%, p=0.01 compared to DAC alone). Responses in treatment naïve patients were seen in 3/7 patients with DAC alone, 2/4 patients with DAC/Carbo and 6/6 patients with DAC/ATO. Fitting a logistic regression model comparing treatments accounting for whether the patients had received prior therapy, the DAC/ATO arm showed a statistically higher response rate as compared to DAC alone (p=0.02). Conclusion: In this randomized phase II study, we find that the combination of decitabine and arsenic trioxide is well tolerated and yields a higher response rate compared to decitabine alone or to decitabine/carboplatin. Our results warrant further studies comparing these regimens for patients with MDS and AML. Disclosures Kropf: Teva Pharmaceuticals: Consultancy. Issa:Astex: Consultancy.
- Published
- 2015
- Full Text
- View/download PDF
48. Abstract CT321: First results of a 10-day regimen of SGI-110 (guadecitabine), a second generation hypomethylating agent (HMA) in previously untreated elderly AML who are not candidates for intensive chemotherapy
- Author
-
Casey O'Connell, Nitin Jain, Gail J. Roboz, Raoul Tibes, Woonbok Chung, Todd L. Rosenblat, Patricia Kropf, Xiang Yao Su, Pietro Taverna, Mohammad Azab, Jean Pierre J. Issa, Wendy Stock, Hagop M. Kantarjian, David A. Rizzieri, Sue Naim, Ellen Richie, Nikola A. Podoltsev, Elizabeth A. Griffiths, and Katherine Walsh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Regimen ,Guadecitabine ,Hypomethylating agent ,business.industry ,Internal medicine ,Medicine ,Intensive chemotherapy ,business ,Intensive care medicine - Abstract
Background: SGI-110 (guadecitabine) is a novel HMA that prolongs in vivo exposure of the active moiety decitabine following subcutaneous (SC) administration. We previously reported guadecitabine clinical activity in previously untreated elderly AML with Overall Complete Response (CR+CRi+CRp) of 55% using a standard 5-day regimen (Yee et al, 2014). Here we report the first results of a more intensive 10-day regimen in the same patient population. Methods: Elderly AML patients (≥ 65y) with at least one of the following characteristics were enrolled: age ≥ 75y; poor Performance Status (PS) ≥ 2; significant comorbidities particularly cardiopulmonary dysfunction; poor risk cytogenetics; or secondary AML. Guadecitabine 60 mg/m2/d SC was administered for 10 days (Days 1-5; and 8-12) for the first 1-2 cycles followed by the 5-day regimen (Days 1-5) for subsequent cycles. Cycles were scheduled every 28 days. Primary endpoint was Overall Complete Response (Overall CR). Secondary endpoints included Overall Survival, Safety including all-cause 30-day and 60-day early mortality, and global maximum DNA demethylation from baseline by LINE-1 assay. Results: Fifty two patients were treated with median age 77 (range 66-92) including 40 (77%) ≥75y; 34 (65%) male; 21 (40%) PS ≥ 2; 19 (36%) poor risk cytogenetics; and 13 (25%) secondary AML. The median bone marrow blasts percentage was 49% (range 16-98%). All patients had a minimum follow up of 3 months; 26 (50%) patients remain on treatment at the time of the analysis. Overall CR was observed in 24 patients (46%):14 CR (27%), 8 CRi (15%), and 2 CRp (4%). There was no significant difference between median DNA demethylation in responders (- 27%) and non-responders (-28%). Early all-cause 30-day and 60-day mortality occurred in 2 (4%) and 10 (19%) patients respectively. The most common Grade ≥3 Adverse Events considered by investigators to be drug-related were febrile neutropenia (33%), thrombocytopenia (27%), neutropenia (21%), anemia (15%), bacteremia (10%), and pneumonia (6%). Conclusions: Administration of guadecitabine as a more intense 10-day regimen for the first 1-2 cycles was active with acceptable toxicity. However, and although it was not a randomized comparison, the 10-day regimen did not result in higher rate of Overall CR in previously untreated AML than what was previously reported for the 5-day regimen. A Phase III randomized clinical trial in previously untreated AML patients not considered candidates for intensive chemotherapy has been initiated with the 5-day regimen of guadecitabine. Reference: Yee K, Daver N, Kropf P, et al: European Hematology Association Meeting, June 12-15, 2104; Milan, Italy. Abstract S647. Citation Format: Gail Roboz, Hagop Kantarjian, Patricia Kropf, Ellen Richie, Nitin Jain, Elizabeth Griffiths, Nikola A. Podoltsev, Katherine Walsh, Casey O'Connell, Wendy Stock, David Rizzieri, Raoul Tibes, Todd Rosenblat, Woonbok Chung, Pietro Taverna, Xiang Yao Su, Sue Naim, Mohammad Azab, Jean-Pierre Issa. First results of a 10-day regimen of SGI-110 (guadecitabine), a second generation hypomethylating agent (HMA) in previously untreated elderly AML who are not candidates for intensive chemotherapy. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT321. doi:10.1158/1538-7445.AM2015-CT321
- Published
- 2015
- Full Text
- View/download PDF
49. Abstract 2952: Determinants of hypomethylation and clinical responses in relapsed/refractory AML patients treated with SGI-110, a novel hypomethylating agent in a phase 1/2 study
- Author
-
Pietro Taverna, Hagop M. Kantarjian, John Lyons, Patricia Kropf, Jean Pierre J. Issa, Yong Hao, Woonbok Chung, and Mohammad Azab
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Cancer ,Decitabine ,Phases of clinical research ,medicine.disease ,Gastroenterology ,Therapeutic index ,Oncology ,Hypomethylating agent ,Refractory ,Internal medicine ,Relapsed refractory ,medicine ,business ,medicine.drug ,Demethylation - Abstract
Introduction: SGI-110 is a dinucleotide of decitabine and deoxyguanosine and a novel subcutaneous (SQ) hypomethylating agent. In a previous Phase 1 (dose escalation) study, we found in relapsed/refractory (r/r) AML patients who were treated with SGI-110 (36 mg/m2-125 mg/m2) subcutaneously (SQ) daily for 5 days a correlation between low LINE-1 demethylation induction, a three gene expression classifier score (low CDA, low P15 and high DNMT3B) and resistance to SGI-110. Here, we analyzed r/r AML patients (n = 122) from Phase 1/2 studies treated at pharmacologically effective doses of SGI-110 looking for determinants of hypomethylation and response. Methods: Phase 1 patients with r/r AML (n = 27) who were treated at a therapeutic dose range of SGI-110 (35 mg/m2 - 125 mg/m2) by SQ daily for 5 days. Phase 2 study r/r AML patients received 60 mg/m2 SQ daily for 5 days (n = 22), 90 mg/m2 SQ daily for 5 days (n = 25) and 60 mg/m2 SQ daily for 10 days (n = 48). Global DNA methylation at pre/post treatment was estimated by bisulfite-pyrosequencing for the LINE-1 repetitive sequence. We also examined expression of a panel of genes (CDA, P15, P21, DNMT3B, DNMT3A, DNMT1, and CTCF) at baseline by quantitative RT-PCR. Results: We analyzed samples from 122 patients with r/r AML. Median age was 59.6 (range, 23-86), 75 were males (61.5%). Overall, peak LINE-1 demethylation generally occurred on day 8 after daily x 5 treatment, or on day 8 or 15 after daily x 10 treatment. In individual patients, peak LINE-1 demethylation ranged from +4.9% to -56.3%. In 122 r/r AML patients, 28 showed overall remission (23.0%, 15 CR and 13 CRi/CRp). Unsupervised clustering by expression of a panel of genes at baseline grouped the patients into two clusters: A (N = 95, response rate = 29.5%) and B (N = 27, response rate = 0%). Cluster B is characterized by high DNMT3b expression, low P15 expression, low CDA expression (average Z-score 1.44 ± 0.26 in cluster B compared to -1.26 ± 0.14 in clusters A, p Conclusions: In a phase 1/2 study of SGI-110, we identified in r/r AML patients a gene expression signature (high DNMT3B, low P15, and low CDA) associated with reduced demethylation and resistance to SGI-110 and we found strong trends for associations between demethylation and response. Citation Format: Woonbok Chung, Pietro Taverna, John Lyons, Yong Hao, Mohammad Azab, Hagop Kantarjian, Patricia Kropf, Jean-Pierre Issa. Determinants of hypomethylation and clinical responses in relapsed/refractory AML patients treated with SGI-110, a novel hypomethylating agent in a phase 1/2 study. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2952. doi:10.1158/1538-7445.AM2015-2952
- Published
- 2015
- Full Text
- View/download PDF
50. Burden of Tyrosine Kinase Inhibitor Failure in Patients with Chronic Myeloid Leukemia
- Author
-
Gisoo Barnes, Patricia Kropf, primary
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.