637 results on '"Michael S Gordon"'
Search Results
2. 756 First-in-human, open-label, multicenter, phase 1 clinical study to evaluate safety, tolerability, pharmacokinetics and pharmacodynamics of anti Siglec-15 PYX-106 in subjects with advanced solid tumors
- Author
-
Bin Zhang, Marsha Crochiere, Anthony W Tolcher, Randy Sweis, Kartik Sehgal, Michael S Gordon, Shui He, Jason Henry, Alexander I Spira, Shiraj Sen, Sandip P Patel, Sondra Smyrnios, and Dipali Unadkat
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. 692 COMMANDER-001: safety data from a phase I/II dose escalation/expansion study of SQZ-eAPC-HPV, a cell-based mRNA therapeutic cancer vaccine for HPV16+ solid tumors
- Author
-
Antonio Jimeno, Michael S Gordon, Howard Bernstein, Scott Loughhead, Jong Chul Park, Justin C Moser, Wade T Iams, Meredith Pelster, Kerry J Rodabaugh, Julia Jennings, Nathan Miselis, Marshelle S Warren, Victoria Villaflor, and Melinda Morrison
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
4. Avelumab for platinum-ineligible/refractory recurrent and/or metastatic squamous cell carcinoma of the head and neck: phase Ib results from the JAVELIN Solid Tumor trial
- Author
-
Nicolas Penel, Ulrich Keilholz, Ani Balmanoukian, Manish R Patel, Sanjay Goel, Keun-Wook Lee, Alain Mita, Christophe Le Tourneau, Deborah J Wong, Patrick Schöffski, Marcis Bajars, Amaury Daste, Hans Juergen Grote, Dongli Zhou, Michael S Gordon, Martin Gutierrez, Damien Vansteene, and Joël Guigay
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
5. Avelumab as second-line therapy for metastatic, platinum-treated urothelial carcinoma in the phase Ib JAVELIN Solid Tumor study: 2-year updated efficacy and safety analysis
- Author
-
James L Gulley, Manish R Patel, Keun-Wook Lee, Luc Dirix, Mary Ruisi, Jeffrey R Infante, Patrick Schöffski, Alain Ravaud, Ding Wang, Andrea B Apolo, John A Ellerton, Manish Agrawal, Michael S Gordon, Raid Aljumaily, Theodore Gourdin, Matthew H Taylor, Juliane Manitz, and Gregory Pennock
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Anti-programmed cell death ligand 1 (PD-L1)/programmed cell death 1 antibodies have shown clinical activity in platinum-treated metastatic urothelial carcinoma, resulting in regulatory approval of several agents, including avelumab (anti-PD-L1). We report ≥2-year follow-up data for avelumab treatment and exploratory subgroup analyses in patients with urothelial carcinoma.Methods Patients with previously treated advanced/metastatic urothelial carcinoma, pooled from two cohorts of the phase Ib JAVELIN Solid Tumor trial, received avelumab 10 mg/kg every 2 weeks until disease progression, unacceptable toxicity or withdrawal. End points included best overall response and progression-free survival (PFS) per RECIST V.1.1, overall survival (OS) and safety. Post hoc analyses included objective response rates (ORRs) in subgroups defined by established high-risk/poor-prognosis characteristics and association between time to response and outcome.Results 249 patients received avelumab; efficacy was assessed in 242 postplatinum patients. Median follow-up was 31.9 months (range 24–43), and median treatment duration was 2.8 months (range 0.5–42.8). The confirmed ORR was 16.5% (95% CI 12.1% to 21.8%; complete response in 4.1% and partial response in 12.4%). Median duration of response was 20.5 months (95% CI 9.7 months to not estimable). Median PFS was 1.6 months (95% CI 1.4 to 2.7 months) and the 12-month PFS rate was 16.8% (95% CI 11.9% to 22.4%). Median OS was 7.0 months (95% CI 5.9 to 8.5 months) and the 24-month OS rate was 20.1% (95% CI 15.2% to 25.4%). In post hoc exploratory analyses, avelumab showed antitumor activity in high-risk subgroups, including elderly patients and those with renal insufficiency or upper tract disease; ORRs were numerically lower in patients with liver metastases or low albumin levels. Objective response achieved by 3 months versus later was associated with longer OS (median not reached (95% CI 18.9 months to not estimable) vs 7.1 months (95% CI 5.2 to 9.0 months)). Safety findings were consistent with previously reported 6-month analyses.Conclusions After ≥2 years of follow-up, avelumab showed prolonged efficacy and acceptable safety in patients with platinum-treated advanced/metastatic urothelial carcinoma, including high-risk subgroups. Survival appeared longer in patients who responded within 3 months. Long-term safety findings were consistent with earlier reports with avelumab treatment in this patient population.
- Published
- 2020
- Full Text
- View/download PDF
6. Pandemic Procedures: Adapting Problem-Solving Court (PSC) Operations and Treatment Protocols During COVID-19
- Author
-
Lindsay R. Smith, Fanni Faragó, James C. Witte, Thomas Blue, Michael S. Gordon, and Faye S. Taxman
- Subjects
Psychiatry and Mental health ,Health (social science) ,Public Health, Environmental and Occupational Health ,Medicine (miscellaneous) - Abstract
With an ongoing pandemic claiming hundreds of lives a day, it is unclear how COVID-19 has affected court operations, particularly problem-solving courts (PSCs) which have goals rooted in rehabilitation for participants in their programs. Even with practical recommendations from national organizations directing courts on how to manage COVID-19, whether and how PSCs met the needs of PSC participants during this time is underexplored. This study, drawn from a larger national study using a survey of PSC coordinators, examines the COVID-19 responses of PSCs to remain safely operational for participants. A sub-sample of survey respondents (n = 82 PSC coordinators) detailed how the COVID-19 pandemic led to changes to their court and treatment operations amidst the constraints of the pandemic. The courts’ shifts in policy and practice have important impacts for court participants’ treatment retention and success in the PSC program, and these shifts need more in-depth research in the future.
- Published
- 2022
7. Supplementary Figure S2 from A Phase I Study of LY3009120, a Pan-RAF Inhibitor, in Patients with Advanced or Metastatic Cancer
- Author
-
David S. Hong, Ramon V. Tiu, Michael S. Gordon, Ilaria Conti, Karim A. Benhadji, Sheng-Bin Peng, James R. Henry, Michael D. Kaufman, Daniel L. Flynn, KrisAnne Crowe, Andrew E. Schade, Danni Yu, Melinda D. Willard, Amanda Sykes, Ling Gao, Wei Zhang, Michael J. Millward, Jordi Rodon Ahnert, Geoffrey I. Shapiro, Keith T. Flaherty, Antoine Hollebecque, and Ryan J. Sullivan
- Abstract
Supplementary Figure S2 shows two cases of patients treated at the RP2D. IHC at Clarient for p-ERK, p27 and Ki-67 were assessed for PD effect using monoclonal rabbit phospho-p44/42 MAPK (ERK1/2), 20G11 Clone; monoclonal mouse anti-Ki67, MIB-1 clone; and monoclonal mouse anti-p27, SX53G8 clone. In (A), the patient is a 56-year old female patient with rectal neuroendocrine cancer who is KRAS and BRAF wild-type. No differences in staining for the 3 biomarkers were observed when baseline and Day 28 samples were compared. In (B), the patient is a 26-year old male patient with hepatocellular carcinoma with unknown mutational status. No differences in staining for Ki-67 or p27 were observed when baseline and Day 28 samples were compared. The p-ERK staining is poor quality and not interpretable. Abbreviations: IHC, immunohistochemistry; PD, pharmacodynamics; RP2D, recommended phase 2 dose
- Published
- 2023
8. Supplementary Material from Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAFV600E-Mutant Colorectal Cancer
- Author
-
Eric Van Cutsem, Fatima Rangwala, Bijoyesh Mookerjee, Severine Bettinger, Savina Jaeger, Jan C. Brase, A. Scott Jung, Filip De Vos, Yves Humblet, Peter J. O'Dwyer, Rona Yaeger, Josep Tabernero, Michael S. Gordon, Kei Muro, Gary Middleton, Salvatore Siena, Autumn J. McRee, Antoine Hollebecque, Johanna C. Bendell, Takayuki Yoshino, Jan H.M. Schellens, Chloe E. Atreya, Thierry André, and Ryan B. Corcoran
- Abstract
Supplementary Material
- Published
- 2023
9. Supplementary Figure 1 from Phase Ia Study of Anti-NaPi2b Antibody–Drug Conjugate Lifastuzumab Vedotin DNIB0600A in Patients with Non–Small Cell Lung Cancer and Platinum-Resistant Ovarian Cancer
- Author
-
Howard A. Burris, Eric W. Humke, Daniel Maslyar, Anjali Vaze, Eva Schuth, Vanessa Lemahieu, Stephanie Royer-Joo, Katie Wood, Kedan Lin, Jian Xu, Divya Samineni, Randall C. Dere, Robert Kahn, YounJeong Choi, David S. Shames, Yulei Wang, Valerie Westcott, Julie Cordova, David R. Spigel, Joan H. Schiller, Maria Martinez Garcia, Enriqueta Felip, Miguel Martín, Sarah B. Goldberg, Michael S. Gordon, Jeffrey R. Infante, and David E. Gerber
- Abstract
Supplementary Figure S1. CA125 response to lifastuzumab vedotin in ovarian cancer patients. All patients with radiographic responses also had {greater than or equal to}50 decrease in CA125 from baseline levels.
- Published
- 2023
10. Supplementary Appendix from Phase Ib Study of Binimetinib with Paclitaxel in Patients with Platinum-Resistant Ovarian Cancer: Final Results, Potential Biomarkers, and Extreme Responders
- Author
-
Carol A. Aghajanian, Vicky Makker, Darragh F. Halpenny, Gwendolyn Cody, Wael Harb, Michael S. Gordon, Kathleen N. Moore, and Rachel N. Grisham
- Abstract
Table S1. Plasma pharmacokinetic sampling times Table S2. Identified Alterations, Listed by Patient Table S3. Adverse events reported in {greater than or equal to}10% of patients (safety cohort) Table S4. Grade 3 to 5 adverse events reported in >1 patient (safety cohort) Table S5. Evaluation of Best Overall Response in Patients with Initial Measurable Disease and Evaluable by CA-125 Figure S1. Geometric mean (standard deviation) plasma concentrations of binimetinib (top) and its metabolite AR00426032 (bottom) on cycle 1 days 7, 8, and 15: semi-logarithmic scale (pharmacokinetic set [30 mg BID binimetinib continuous]) Figure S2. Geometric mean (standard deviation) plasma concentrations of binimetinib (top) and its metabolite AR00426032 (bottom) on cycle 1 days 7, 8, and 15: semi-logarithmic scale (pharmacokinetic set [45 mg BID binimetinib continuous]) Figure S3. Geometric mean (standard deviation) plasma concentrations of binimetinib (top) and its metabolite AR00426032 (bottom) on cycle 1 days 1, 8, and 15: semi-logarithmic scale (pharmacokinetic set [45 mg BID binimetinib intermittent]) Figure S4. Representative CT Resolution of Implant in a Complete Responder
- Published
- 2023
11. Supplementary Methods and Tables 1 - 3 from Safety, Pharmacokinetics, and Pharmacodynamics of AMG 102, a Fully Human Hepatocyte Growth Factor–Neutralizing Monoclonal Antibody, in a First-in-Human Study of Patients with Advanced Solid Tumors
- Author
-
Lia Gore, Min Zhu, Lucy Yan, Kelly S. Oliner, Ian M. Leitch, Paula Kaplan-Lefko, Hongjie Deng, Angela Coxon, Teresa L. Burgess, Daniel Branstetter, Darrin M. Beaupre, Abraham Anderson, S. Gail Eckhardt, David S. Mendelson, Christopher J. Sweeney, and Michael S. Gordon
- Abstract
PDF file - 73K
- Published
- 2023
12. Table S1 from A Phase I Study of ASTX660, an Antagonist of Inhibitors of Apoptosis Proteins, in Adults with Advanced Cancers or Lymphoma
- Author
-
Anthony W. Tolcher, Simone Jueliger, Xiang Yao Su, Aram Oganesian, Harold Keer, Roberta Ferraldeschi, Alain C. Mita, Michael S. Gordon, Kyriakos P. Papadopoulos, Patricia M. LoRusso, and Monica M. Mita
- Abstract
Table S1 shows dose levels of ASTX660 by cohort
- Published
- 2023
13. Data from Phase I Studies of CBP501, a G2 Checkpoint Abrogator, as Monotherapy and in Combination with Cisplatin in Patients with Advanced Solid Tumors
- Author
-
Sunil Sharma, Takumi Kawabe, Daniel Von Hoff, Donald W. Kufe, Ernesto Wasserman, Scott Slough, David Weaver, William E. Pierceall, Hitoshi Sato, William Sutherland, Cristian Fernandez, Glen J. Weiss, Bruno R. Bastos, Nicholas J. Vogelzang, David S. Mendelson, Mitesh J. Borad, Joseph Paul Eder, Bryan Y. Wong, Michael S. Gordon, Raoul Tibes, and Geoffrey I. Shapiro
- Abstract
Purpose: Two phase I dose-escalation studies were conducted to determine the maximum tolerated dose (MTD) and safety profile of the G2 checkpoint abrogator CBP501, as a single agent and in combination with cisplatin.Experimental Design: Patients with advanced solid tumors were treated with CBP501 alone (D1/D8/D15, q4w, from 0.9 mg/m2), or with cisplatin (both on D1, q3w, from 3.6 mg/m2 CBP501, 50 mg/m2 cisplatin). Dose escalation proceeded if dose-limiting toxicity (DLT) was observed in 1 or less of 3 to 6 patients; CBP501 dose increments were implemented according to the incidence of toxicity. MTD was determined from DLTs occurring during the first two cycles.Results: In the combination study, the DLT was a histamine-release syndrome (HRS) occurring 10 to 60 minutes after initiating infusion that was attenuated by prophylaxis comprising dexamethasone, diphenhydramine, ranitidine, and loratadine. The MTD was 25 mg/m2 CBP501 and 75 mg/m2 cisplatin, with two patients at the highest dose (36.4 mg/m2 CBP501, 75 mg/m2 cisplatin) experiencing grade 3 HRS. The only DLT with monotherapy was transient G3 rise of troponin in one patient. Grade 3 to 4 treatment–related events were rare. Promising activity was observed with CBP501/cisplatin, mainly in ovarian and mesothelioma patients who had previously progressed on platinum-containing regimens. Among ovarian cancer patients, low expression of DNA repair proteins was associated with partial response or stable disease.Conclusions: CBP501 is well tolerated in patients as monotherapy and with cisplatin. At the recommended phase II dose (RP2D), the combination is feasible and HRS manageable with prophylaxis. Evidence of antitumor activity was observed in platinum-resistant patients. Clin Cancer Res; 17(10); 3431–42. ©2011 AACR.
- Published
- 2023
14. Supplementary Figure 1 from Safety, Pharmacokinetics, Pharmacodynamics, and Antitumor Activity of Dalantercept, an Activin Receptor–like Kinase-1 Ligand Trap, in Patients with Advanced Cancer
- Author
-
Sunil Sharma, Matthew L. Sherman, Kenneth M. Attie, Ty McClure, Yijun Yang, Amelia E. Pearsall, Dawn Wilson, Carolyn H. Condon, Neeraj Agarwal, Gerard C. Blobe, David S. Mendelson, Suzanne F. Jones, Herbert I. Hurwitz, Michael S. Gordon, and Johanna C. Bendell
- Abstract
PDF file - 7635K, Mean (plus-minus standard error) serum concentration of dalantercept versus time after first dose for each dose level.
- Published
- 2023
15. Data from BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer
- Author
-
Ryan B. Corcoran, Eduard Gasal, Fatima Rangwala, Jan C. Brase, John M. Millholland, Eric Van Cutsem, Filip De Vos, Peter J. O'Dwyer, Rona Yaeger, Josep Tabernero, Michael S. Gordon, Salvatore Siena, Autumn J. McRee, Antoine Hollebecque, Johanna C. Bendell, Takayuki Yoshino, Jan H.M. Schellens, Chloe E. Atreya, Thierry André, Catarina D. Campbell, Yiqun Yang, and Gary Middleton
- Abstract
Purpose:The influence of the transcriptional and immunologic context of mutations on therapeutic outcomes with targeted therapy in cancer has not been well defined. BRAF V600E–mutant (BM) colorectal cancer comprises two main transcriptional subtypes, BM1 and BM2. We sought to determine the impact of BM subtype, as well as distinct biological features of those subtypes, on response to BRAF/MEK/EGFR inhibition in patients with colorectal cancer.Patients and Methods:Paired fresh tumor biopsies were acquired at baseline and on day 15 of treatment from all consenting patients with BM colorectal cancer enrolled in a phase II clinical trial of dabrafenib, trametinib, and panitumumab. For each sample, BM subtype, cell cycle, and immune gene signature expression were determined using RNA-sequencing (RNA-seq), and a Cox proportional hazards model was applied to determine association with progression-free survival (PFS).Results:Confirmed response rates, median PFS, and median overall survival (OS) were higher in BM1 subtype patients compared with BM2 subtype patients. Evaluation of immune contexture identified greater immune reactivity in BM1, whereas cell-cycle signatures were more highly expressed in BM2. A multivariate model of PFS incorporating BM subtype plus immune and cell-cycle signatures revealed that BM subtype encompasses the majority of the effect.Conclusions:BM subtype is significantly associated with the outcome of combination dabrafenib, trametinib, and panitumumab therapy and may serve as a standalone predictive biomarker beyond mutational status. Our findings support a more nuanced approach to targeted therapeutic decisions that incorporates assessment of transcriptional context.
- Published
- 2023
16. Figure S1 from A Phase I Study of ASTX660, an Antagonist of Inhibitors of Apoptosis Proteins, in Adults with Advanced Cancers or Lymphoma
- Author
-
Anthony W. Tolcher, Simone Jueliger, Xiang Yao Su, Aram Oganesian, Harold Keer, Roberta Ferraldeschi, Alain C. Mita, Michael S. Gordon, Kyriakos P. Papadopoulos, Patricia M. LoRusso, and Monica M. Mita
- Abstract
Figure S1 shows ASTX660 dose proportionality-power model analyses on AUC0-24 and Cmax by formulation.
- Published
- 2023
17. Supplementary Data from BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer
- Author
-
Ryan B. Corcoran, Eduard Gasal, Fatima Rangwala, Jan C. Brase, John M. Millholland, Eric Van Cutsem, Filip De Vos, Peter J. O'Dwyer, Rona Yaeger, Josep Tabernero, Michael S. Gordon, Salvatore Siena, Autumn J. McRee, Antoine Hollebecque, Johanna C. Bendell, Takayuki Yoshino, Jan H.M. Schellens, Chloe E. Atreya, Thierry André, Catarina D. Campbell, Yiqun Yang, and Gary Middleton
- Abstract
Supplementary Data
- Published
- 2023
18. Data from Safety, Pharmacokinetics, Pharmacodynamics, and Antitumor Activity of Dalantercept, an Activin Receptor–like Kinase-1 Ligand Trap, in Patients with Advanced Cancer
- Author
-
Sunil Sharma, Matthew L. Sherman, Kenneth M. Attie, Ty McClure, Yijun Yang, Amelia E. Pearsall, Dawn Wilson, Carolyn H. Condon, Neeraj Agarwal, Gerard C. Blobe, David S. Mendelson, Suzanne F. Jones, Herbert I. Hurwitz, Michael S. Gordon, and Johanna C. Bendell
- Abstract
Purpose: The angiogenesis inhibitor dalantercept (formerly ACE-041) is a soluble form of activin receptor–like kinase-1 (ALK1) that prevents activation of endogenous ALK1 by bone morphogenetic protein-9 (BMP9) and BMP10 and exhibits antitumor activity in preclinical models. This first-in-human study of dalantercept evaluated its safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity in adults with advanced solid tumors.Experimental Design: Patients in dose-escalating cohorts received dalantercept subcutaneously at one of seven dose levels (0.1–4.8 mg/kg) every 3 weeks until disease progression. Patients in an expansion cohort received dalantercept at 0.8 or 1.6 mg/kg every 3 weeks until disease progression.Results: In 37 patients receiving dalantercept, the most common treatment-related adverse events were peripheral edema, fatigue, and anemia. Edema and fluid retention were dose-limiting toxicities and responded to diuretic therapy. No clinically significant, treatment-related hypertension, proteinuria, gross hemorrhage, or gastrointestinal perforations were observed. One patient with refractory squamous cell cancer of the head and neck had a partial response, and 13 patients had stable disease according to RECISTv1.1, eight of whom had prolonged periods (≥12 weeks) of stable disease. Correlative pharmacodynamic markers included tumor metabolic activity and tumor blood flow, which decreased from baseline in 63% and 82% of evaluable patients, respectively, and telangiectasia in eight patients.Conclusion: Dalantercept was well-tolerated at doses up to 1.6 mg/kg, with a safety profile distinct from inhibitors of the VEGF pathway. Dalantercept displayed promising antitumor activity in patients with advanced refractory cancer, and multiple phase II studies are underway. Clin Cancer Res; 20(2); 480–9. ©2013 AACR.
- Published
- 2023
19. Data from Phase Ia Study of Anti-NaPi2b Antibody–Drug Conjugate Lifastuzumab Vedotin DNIB0600A in Patients with Non–Small Cell Lung Cancer and Platinum-Resistant Ovarian Cancer
- Author
-
Howard A. Burris, Eric W. Humke, Daniel Maslyar, Anjali Vaze, Eva Schuth, Vanessa Lemahieu, Stephanie Royer-Joo, Katie Wood, Kedan Lin, Jian Xu, Divya Samineni, Randall C. Dere, Robert Kahn, YounJeong Choi, David S. Shames, Yulei Wang, Valerie Westcott, Julie Cordova, David R. Spigel, Joan H. Schiller, Maria Martinez Garcia, Enriqueta Felip, Miguel Martín, Sarah B. Goldberg, Michael S. Gordon, Jeffrey R. Infante, and David E. Gerber
- Abstract
Purpose:This phase I trial assessed the safety, tolerability, and preliminary antitumor activity of lifastuzumab vedotin (LIFA), an antibody–drug conjugate of anti-NaPi2b mAb (MNIB2126A) and a potent antimitotic agent (monomethyl auristatin E).Patients and Methods:LIFA was administered to patients with non–small cell lung cancer (NSCLC) and platinum-resistant ovarian cancer (PROC), once every 3 weeks, by intravenous infusion. The starting dose was 0.2 mg/kg in this 3+3 dose-escalation design, followed by cohort expansion at the recommended phase II dose (RP2D).Results:Overall, 87 patients were treated at doses between 0.2 and 2.8 mg/kg. The MTD was not reached; 2.4 mg/kg once every 3 weeks was selected as the RP2D based on overall tolerability profile. The most common adverse events of any grade and regardless of relationship to study drug were fatigue (59%), nausea (49%), decreased appetite (37%), vomiting (32%), and peripheral sensory neuropathy (29%). Most common treatment-related grade ≥3 toxicities among patients treated at the RP2D (n = 63) were neutropenia (10%), anemia (3%), and pneumonia (3%). The pharmacokinetic profile was dose proportional. At active doses ≥1.8 mg/kg, partial responses were observed in four of 51 (8%) patients with NSCLC and 11 of 24 (46%) patients with PROC per RECIST. All RECIST responses occurred in patients with NaPi2b-high by IHC. The CA-125 biomarker assessed for patients with PROC dosed at ≥1.8 mg/kg showed 13 of 24 (54%) had responses (≥50% decline from baseline).Conclusions:LIFA exhibited dose-proportional pharmacokinetics and an acceptable safety profile, with encouraging activity in patients with PROC at the single-agent RP2D of 2.4 mg/kg.
- Published
- 2023
20. Data from A Phase I Study of ASTX660, an Antagonist of Inhibitors of Apoptosis Proteins, in Adults with Advanced Cancers or Lymphoma
- Author
-
Anthony W. Tolcher, Simone Jueliger, Xiang Yao Su, Aram Oganesian, Harold Keer, Roberta Ferraldeschi, Alain C. Mita, Michael S. Gordon, Kyriakos P. Papadopoulos, Patricia M. LoRusso, and Monica M. Mita
- Abstract
Purpose:This first-in-human, phase I study evaluated ASTX660, an oral, small-molecule antagonist of cellular/X-linked inhibitors of apoptosis proteins in patients with advanced solid tumors or lymphoma.Patients and Methods:ASTX660 was administered orally once daily on a 7-day-on/7-day-off schedule in a 28-day cycle. Dose escalation followed a standard 3+3 design to determine the MTD and recommended phase II dose (RP2D). Dose expansion was conducted at the RP2D.Results:Forty-five patients received ASTX660 (range 15–270 mg/day). Dose-limiting toxicity of grade 3 increased lipase with or without increased amylase occurred in 3 patients at 270 mg/day and 1 patient at 210 mg/day. The MTD was determined to be 210 mg/day and the RP2D 180 mg/day. Common treatment-related adverse events included fatigue (33%), vomiting (31%), and nausea (27%). Grade ≥3 treatment-related adverse events occurred in 7 patients, most commonly anemia (13%), increased lipase (11%), and lymphopenia (9%). ASTX660 was rapidly absorbed, with maximum concentration achieved at approximately 0.5–1.0 hour. An approximately 2-fold accumulation in AUC exposures was observed on day 7 versus 1. ASTX660 suppressed cellular inhibitor of apoptosis protein-1 in peripheral blood mononuclear cells, which was maintained into the second cycle beyond the off-therapy week at the 180-mg/day RP2D and above. Clinical activity was seen in a patient with cutaneous T-cell lymphoma.Conclusions:ASTX660 demonstrated a manageable safety profile and exhibited evidence of pharmacodynamic and preliminary clinical activity at the 180-mg/day RP2D. The phase II part of the study is ongoing.
- Published
- 2023
21. Supplementary Data from Phase I Studies of CBP501, a G2 Checkpoint Abrogator, as Monotherapy and in Combination with Cisplatin in Patients with Advanced Solid Tumors
- Author
-
Sunil Sharma, Takumi Kawabe, Daniel Von Hoff, Donald W. Kufe, Ernesto Wasserman, Scott Slough, David Weaver, William E. Pierceall, Hitoshi Sato, William Sutherland, Cristian Fernandez, Glen J. Weiss, Bruno R. Bastos, Nicholas J. Vogelzang, David S. Mendelson, Mitesh J. Borad, Joseph Paul Eder, Bryan Y. Wong, Michael S. Gordon, Raoul Tibes, and Geoffrey I. Shapiro
- Abstract
Supplementary Figure S1; Supplementary Tables S1-S2.
- Published
- 2023
22. Supplementary Figure 2 from Phase Ia Study of Anti-NaPi2b Antibody–Drug Conjugate Lifastuzumab Vedotin DNIB0600A in Patients with Non–Small Cell Lung Cancer and Platinum-Resistant Ovarian Cancer
- Author
-
Howard A. Burris, Eric W. Humke, Daniel Maslyar, Anjali Vaze, Eva Schuth, Vanessa Lemahieu, Stephanie Royer-Joo, Katie Wood, Kedan Lin, Jian Xu, Divya Samineni, Randall C. Dere, Robert Kahn, YounJeong Choi, David S. Shames, Yulei Wang, Valerie Westcott, Julie Cordova, David R. Spigel, Joan H. Schiller, Maria Martinez Garcia, Enriqueta Felip, Miguel Martín, Sarah B. Goldberg, Michael S. Gordon, Jeffrey R. Infante, and David E. Gerber
- Abstract
Supplementary Figure S2. Representative NaPi2b ICH images for PROC and NSCLC patients.
- Published
- 2023
23. Data from Safety, Pharmacokinetics, and Pharmacodynamics of AMG 102, a Fully Human Hepatocyte Growth Factor–Neutralizing Monoclonal Antibody, in a First-in-Human Study of Patients with Advanced Solid Tumors
- Author
-
Lia Gore, Min Zhu, Lucy Yan, Kelly S. Oliner, Ian M. Leitch, Paula Kaplan-Lefko, Hongjie Deng, Angela Coxon, Teresa L. Burgess, Daniel Branstetter, Darrin M. Beaupre, Abraham Anderson, S. Gail Eckhardt, David S. Mendelson, Christopher J. Sweeney, and Michael S. Gordon
- Abstract
Purpose: The aims were to assess the safety, pharmacokinetics, maximum tolerated dose, and antitumor activity of AMG 102, a fully human hepatocyte growth factor/scatter factor (HGF/SF)–neutralizing monoclonal antibody, in patients with solid tumors.Experimental Design: Patients (N = 40) with refractory advanced solid tumors were enrolled into six sequential dose-escalation cohorts (0.5, 1, 3, 5, 10, or 20 mg/kg AMG 102 i.v. every 2 weeks) and a dose-expansion cohort (20 mg/kg AMG 102 every 2 weeks). Safety, anti–AMG 102 antibody formation, pharmacokinetics, tumor response, and exploratory biomarkers were assessed.Results: AMG 102 was well tolerated up to the planned maximum dose of 20 mg/kg, and the maximum tolerated dose was not reached. Treatment-related adverse events were generally mild and included fatigue (13%), constipation (8%), nausea (8%), vomiting (5%), anorexia (5%), myalgia (5%), and hypertension (5%). Two patients experienced dose-limiting toxicities: one patient (0.5 mg/kg cohort) experienced grade 3 hypoxia and grade 3 dyspnea and one patient (1 mg/kg cohort) experienced grade 3 upper gastrointestinal hemorrhage. No anti–AMG 102 antibodies were detected, and AMG 102 had linear pharmacokinetics within the dose range investigated. Sixteen of 23 (70%) evaluable patients had a best response of stable disease with progression-free survival ranging from 7.9 to 40 weeks. Circulating levels of the biomarker HGF/SF (bound and unbound) increased in a dose-dependent manner, whereas soluble c-Met concentrations were generally similar across doses.Conclusions: AMG 102 is safe and well tolerated, has a favorable pharmacokinetic profile, and will be further investigated as a monotherapy and in combination with other agents. Clin Cancer Res; 16(2); 699–710
- Published
- 2023
24. Supplementary Tables from Phase Ia Study of Anti-NaPi2b Antibody–Drug Conjugate Lifastuzumab Vedotin DNIB0600A in Patients with Non–Small Cell Lung Cancer and Platinum-Resistant Ovarian Cancer
- Author
-
Howard A. Burris, Eric W. Humke, Daniel Maslyar, Anjali Vaze, Eva Schuth, Vanessa Lemahieu, Stephanie Royer-Joo, Katie Wood, Kedan Lin, Jian Xu, Divya Samineni, Randall C. Dere, Robert Kahn, YounJeong Choi, David S. Shames, Yulei Wang, Valerie Westcott, Julie Cordova, David R. Spigel, Joan H. Schiller, Maria Martinez Garcia, Enriqueta Felip, Miguel Martín, Sarah B. Goldberg, Michael S. Gordon, Jeffrey R. Infante, and David E. Gerber
- Abstract
Supplementary Table S1. Adverse events regardless of relationship to study drug occurring in 10 or more patients Supplementary Table S2. Adverse events grades 3-5 regardless of relationship to study drug occurring in 2 or more patients Supplementary Table S3. Pulmonary adverse events by frequency of occurrence in all patients regardless of relationship to study drug Supplementary Table S4. Pulmonary adverse events of NCI-CTCAE grade 3 or higher by frequency of occurrence in all patients regardless of relationship to study drug Supplementary Table S5. Mean (standard deviation) of the pharmacokinetic parameters for antibody-conjugated MMAE in plasma Supplementary Table S6. Mean (standard deviation) of the pharmacokinetic parameters for total antibody in plasma Supplementary Table S7. Mean (standard deviation) of the pharmacokinetic parameters for unconjugated MMAE in plasma Supplementary Table S8. Response outcomes and duration
- Published
- 2023
25. Data from Phase Ib Study of Binimetinib with Paclitaxel in Patients with Platinum-Resistant Ovarian Cancer: Final Results, Potential Biomarkers, and Extreme Responders
- Author
-
Carol A. Aghajanian, Vicky Makker, Darragh F. Halpenny, Gwendolyn Cody, Wael Harb, Michael S. Gordon, Kathleen N. Moore, and Rachel N. Grisham
- Abstract
Purpose: Epithelial ovarian cancer (EOC) is a molecularly diverse disease. MEK inhibition targets tumors harboring MAPK pathway alterations and enhances paclitaxel-induced apoptosis in EOC. This phase Ib study evaluated the MEK inhibitor binimetinib combined with paclitaxel in patients with platinum-resistant EOC.Patients and Methods: Patients received intravenous weekly paclitaxel with oral binimetinib in three different administration schedules. Outcomes were assessed by RECIST and CGIC CA-125 response criteria. Tumor samples were analyzed using next-generation sequencing.Results: Thirty-four patients received ≥1 binimetinib dose. A 30-mg twice-a-day continuous or 45-mg twice-a-day intermittent binimetinib dose was deemed the recommended phase II dose (RP2D) in combination with 80 mg/m2 i.v. weekly paclitaxel. Rate of grade 3/4 adverse events was 65%. The best overall response rate was 18%—one complete (CR) and four partial responses (PR)—among 28 patients with RECIST-measurable disease. Eleven patients achieved stable disease (SD), yielding a clinical benefit rate (CR+PR+SD) of 57%. Response rates, per both RECIST and CA-125 criteria, were highest in the 45-mg twice-a-day continuous cohort and lowest in the 45-mg twice-a-day intermittent cohort. All four evaluable patients with MAPK pathway–altered tumors experienced clinical benefit.Conclusions: The combination of binimetinib and intravenous weekly paclitaxel was tolerable in this patient population. The RP2D of binimetinib in combination with paclitaxel was 30 mg twice a day as a continuous or 45 mg twice a day as an intermittent dose. Although response rates were modest, a higher clinical benefit rate was seen in patients harboring alterations affecting the MAPK pathway. Clin Cancer Res; 24(22); 5525–33. ©2018 AACR.
- Published
- 2023
26. Supplementary Data from Safety, Pharmacokinetics, and Pharmacodynamics of AMG 102, a Fully Human Hepatocyte Growth Factor–Neutralizing Monoclonal Antibody, in a First-in-Human Study of Patients with Advanced Solid Tumors
- Author
-
Lia Gore, Min Zhu, Lucy Yan, Kelly S. Oliner, Ian M. Leitch, Paula Kaplan-Lefko, Hongjie Deng, Angela Coxon, Teresa L. Burgess, Daniel Branstetter, Darrin M. Beaupre, Abraham Anderson, S. Gail Eckhardt, David S. Mendelson, Christopher J. Sweeney, and Michael S. Gordon
- Abstract
Supplementary Data from Safety, Pharmacokinetics, and Pharmacodynamics of AMG 102, a Fully Human Hepatocyte Growth Factor–Neutralizing Monoclonal Antibody, in a First-in-Human Study of Patients with Advanced Solid Tumors
- Published
- 2023
27. Supplementary Figure 2 from Safety, Pharmacokinetics, Pharmacodynamics, and Antitumor Activity of Dalantercept, an Activin Receptor–like Kinase-1 Ligand Trap, in Patients with Advanced Cancer
- Author
-
Sunil Sharma, Matthew L. Sherman, Kenneth M. Attie, Ty McClure, Yijun Yang, Amelia E. Pearsall, Dawn Wilson, Carolyn H. Condon, Neeraj Agarwal, Gerard C. Blobe, David S. Mendelson, Suzanne F. Jones, Herbert I. Hurwitz, Michael S. Gordon, and Johanna C. Bendell
- Abstract
PDF file - 43K, Tumor blood flow response to dalantercept as assessed by DCE-MRI. Best response measured as maximum percent change from baseline in median Ktrans at day 15.
- Published
- 2023
28. Safety and Clinical Activity of Atezolizumab Plus Ipilimumab in Locally Advanced or Metastatic Non–Small Cell Lung Cancer: Results From a Phase 1b Trial
- Author
-
Michael S. Gordon, Todd M. Bauer, Xiaosong Zhang, Fabiola Bene-Tchaleu, Jing Zhu, Edward Cha, and Deborah J. Wong
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Population ,Ipilimumab ,Antibodies, Monoclonal, Humanized ,Atezolizumab ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Lung cancer ,Adverse effect ,education ,Melanoma ,Pneumonitis ,education.field_of_study ,business.industry ,medicine.disease ,Tolerability ,business ,medicine.drug - Abstract
Background : This phase 1b study investigated safety and activity of combined checkpoint inhibition (CPI) with programmed death-ligand 1 (PD-L1) antibody atezolizumab plus cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitor ipilimumab in NSCLC. Patients and Methods : Eligible patients had previously treated locally advanced or metastatic non-small cell lung cancer (NSCLC) or melanoma. A standard 3+3 dose escalation investigated atezolizumab (600-1200 mg IV every 3 weeks) plus ipilimumab starting at 1 mg/kg, administered as a single dose or 4 doses, administered every 3 weeks. The expansion stage included a cohort previously treated with atezolizumab. Patients were monitored for safety and tolerability; response was evaluated every 6 weeks. Results : Twenty-seven patients were enrolled, 4 with melanoma and 23 with NSCLC; here, we focus on data for the NSCLC population. Three of 23 patients (13.0%) received prior CPI. No dose-limiting toxicities were reported during dose escalation; dose expansion occurred with atezolizumab 1200 mg plus 1 cycle of ipilimumab 1 mg/kg. Most common treatment-emergent adverse events (AEs) were dyspnea (39%) and cough (35%); treatment-related grade ≥3 AEs occurred in 11 patients (48%), most frequently pneumonitis (17%) and amylase or lipase elevation (9% each). Six of 23 NSCLC patients (26%) achieved confirmed responses, 5 of whom (25%) were CPI-naive. Median duration of response was 23.0 (95% CI, 3.2-36.9) months overall and 36.9 (95% CI, 2.9-36.9) months in CPI-naive patients. Conclusion : Preliminary efficacy of atezolizumab plus ipilimumab was observed in metastatic NSCLC. The combination had manageable toxicity, with a safety profile consistent with those of the individual agents. MicroAbstract : Dual checkpoint inhibitor therapy with atezolizumab (anti–PD-L1) and ipilimumab (anti–CTLA-4) may improve outcomes in locally advanced or metastatic NSCLC. This phase 1b study investigated atezolizumab plus ipilimumab in 23 patients with previously treated NSCLC. The combination had manageable toxicity and demonstrated preliminary clinical activity. Further elucidation of biomarkers to optimally select patients for checkpoint inhibitor therapy may be required.
- Published
- 2022
29. Phase 1 study to determine the safety and dosing of autologous PBMCs modified to present HPV16 antigens (SQZ- PBMC-HPV) in HLA-A*02+ patients with HPV16+ Solid Tumors
- Author
-
Antonio Jimeno, Joaquina Baranda, Wade T. Iams, Jong Chul Park, Monica Mita, Michael S. Gordon, Matthew Taylor, Neesha Dhani, Alexis D. Leal, Prakash Neupane, Cathy Eng, Oladapo Yeku, Alain Mita, Justin C. Moser, Marcus Butler, Scott M. Loughhead, Julia Jennings, Nathan R. Miselis, Rui-Ru Ji, Nitya Nair, Martin Kornacker, Ricardo F. Zwirtes, Howard Bernstein, and Armon Sharei
- Subjects
Pharmacology ,Oncology ,Pharmacology (medical) - Abstract
Purpose We conducted a dose escalation Phase 1 study of autologous PBMCs loaded by microfluidic squeezing (Cell Squeeze® technology) with HPV16 E6 and E7 antigens (SQZ-PBMC-HPV), in HLA-A*02 + patients with advanced/metastatic HPV16 + cancers. Preclinical studies in murine models had shown such cells resulted in stimulation and proliferation of antigen specific CD8 + cells, and demonstrated antitumor activity. Methods Administration of SQZ-PBMC-HPV was every 3 weeks. Enrollment followed a modified 3 + 3 design with primary objectives to define safety, tolerability, and the recommended Phase 2 dose. Secondary and exploratory objectives were antitumor activity, manufacturing feasibility, and pharmacodynamic evaluation of immune responses. Results Eighteen patients were enrolled at doses ranging from 0.5 × 106to 5.0 × 106live cells/kg. Manufacture proved feasible and required 6live cells/kg with double priming was chosen as the recommended Phase 2 dose. Multiple participants exhibited pharmacodynamic changes consistent with immune responses supporting the proposed mechanism of action for SQZ-PBMC-HPV, including patients previously refractory to checkpoint inhibitors.
- Published
- 2022
30. LB1530. Clinical Benefit of Oral sabizabulin for Hospitalized Adults with CoVID-19 on Supplemental Oxygen
- Author
-
Tara L Gonzales, Paula Skarda, Thomas G Bird, Michael Schnaus, Mitchell Steiner, Alan Skolnick, K Gary Barnette, Michael S Gordon, Eduardo Sprinz, Domingo Rodriguez, Petar Kalaydzhiev, and Georgi Arabadzhiev
- Subjects
Infectious Diseases ,Oncology - Abstract
Background Sabizabulin is an oral, novel microtubule disruptor with dual antiviral and anti-inflammatory activities. A randomized, multicenter placebo-controlled Phase 3 clinical trial was conducted in hospitalized moderate-severe COVID-19 patients at high-risk for acute respiratory distress syndrome (ARDS) and death. Patients were randomized (2:1) to sabizabulin 9mg or placebo oral daily dose (up to 21 days). In a planned interim analysis, sabizabulin treatment resulted in a 55.2% relative reduction in mortality compared to placebo. Methods The primary endpoint was all-cause mortality up to day 60. Key secondary endpoints were days in intensive care unit (ICU), on mechanical ventilation, and in hospital. Randomization was stratified by oxygen requirement at baseline (WHO 4 = supplemental oxygen, WHO 5 = NIV/forced oxygen, WHO 6 = mechanical ventilation). The WHO 4 patients also were required to have at least one comorbidity (Asthma, Chronic Lung Disease, Diabetes, Hypertension, Severe Obesity (BMI ≥40), ≥65 years of age, in a nursing/long-term care facility, or immunocompromised). A post-hoc analysis of the key efficacy outcomes in WHO 4 at baseline patients with a comorbidity was conducted. Results A total of 88 patients classified as WHO 4 with a baseline comorbidity underwent randomization (59 sabizabulin/29 placebo). Baseline characteristics were similar. Sabizabulin treatment resulted in a 22.4 absolute percentage point and 81.2% relative reduction in deaths compared to the placebo (odds ratio 6.22, 95% CI [1.58 to 24.48], p=0.0090). Mortality rate was 5.2% (3 of 58) for sabizabulin versus 27.6% (8 of 29) for placebo. Key secondary endpoints: sabizabulin treatment resulted in relative reductions of 74.7% in days in ICU (p=0.0021), 80.7% in days on mechanical ventilation (p=0.0019), and 39.8% in days in hospital (p=0.0191) vs placebo. Conclusion Statistically and clinically significant reductions in mortality, days in the ICU, on mechanical ventilation, and in the hospital were observed in the sabizabulin treated compared to placebo hospitalized COVID-19 WHO-4 patients with at least one comorbidity suggesting that the antiviral action of sabizabulin contributes early in the prevention of COVID-19 progression to ARDS and death. Disclosures Tara L. Gonzales, MD, Veru Inc.: Employee Mitchell Steiner, MD, Veru, Inc: Board Member|Veru, Inc: Stocks/Bonds K. Gary Barnette, PhD, Veru Inc.: Employee|Veru Inc.: Ownership Interest Michael S. Gordon, MD, Agenus: Grant/Research Support|Arcus: Grant/Research Support|Astex: Grant/Research Support|ATEA: Grant/Research Support|Beigene: Grant/Research Support|Caremission: Ownership Interest|Celldex: Grant/Research Support|Corcept: Grant/Research Support|Daiichi: Grant/Research Support|Deciphera: Grant/Research Support|Endocyte: Grant/Research Support|Forma: Grant/Research Support|FujiFilm: Grant/Research Support|Genentech/Roche: Grant/Research Support|I-MAB Pharma: Grant/Research Support|Imaginab: Advisor/Consultant|Imaginab: Grant/Research Support|Imaging Endpoints: Advisor/Consultant|Incyte: Grant/Research Support|Kinevant: Grant/Research Support|Medelis: Ownership Interest|Medimmune: Grant/Research Support|Morphic Tx: Advisor/Consultant|Nikang: Grant/Research Support|OncoResponse: Grant/Research Support|OnQuality: Advisor/Consultant|Pfizer: Advisor/Consultant|Pfizer: Grant/Research Support|Pionyr: Grant/Research Support|Plexxicon: Grant/Research Support|Qualigen: Advisor/Consultant|RedHill Bio: Grant/Research Support|Revolution Medicine: Grant/Research Support|Riboscience: Grant/Research Support|Seattle Genetics: Grant/Research Support|Serono: Grant/Research Support|SQZ Biotech: Grant/Research Support|Syndax: Grant/Research Support|Theseus: Grant/Research Support|Tolero: Grant/Research Support|Tracon: Grant/Research Support|Vedanta: Grant/Research Support|Veru: Grant/Research Support Eduardo Sprinz, MD; MsC, ScD, Gilead: Advisor/Consultant|Gilead: Board Member|GSK: Advisor/Consultant|GSK: Board Member|GSK: Grant/Research Support|GSK: Honoraria|Janssen: Advisor/Consultant|Janssen: Board Member|Janssen: Honoraria Domingo Rodriguez, n/a, Veru: Stocks/Bonds Domingo Rodriguez, n/a, Veru: Stocks/Bonds.
- Published
- 2022
31. A phase 1 study to assess the safety, pharmacokinetics, and anti-tumor activity of the androgen receptor n-terminal domain inhibitor epi-506 in patients with metastatic castration-resistant prostate cancer
- Author
-
Leah DiMascio, Frank Perabo, Corinne Maurice-Dror, Ronan Le Moigne, Michael S. Gordon, Ulka N. Vaishampayan, Kim N. Chi, Nan Hyung Hong, and Robert B. Montgomery
- Subjects
Pharmacology ,Oncology ,medicine.medical_specialty ,business.industry ,Nausea ,medicine.disease ,Androgen receptor ,Prostate cancer ,Pharmacokinetics ,Tolerability ,Internal medicine ,Vomiting ,Medicine ,Potency ,Pharmacology (medical) ,medicine.symptom ,business ,Adverse effect - Abstract
BACKGROUND EPI-506 is the first of a new class of drugs targeting the N-terminal domain (NTD) of the androgen receptor (AR), potentially overcoming known resistance mechanisms to androgen receptor pathway inhibitors (ARPIs) among men with metastatic castration resistant prostate cancer (mCRPC). METHODS Patients with mCRPC who had progressed on prior ARPI were enrolled in this phase 1 open-label, adaptive 3 + 3 dose escalation study. The primary outcome was safety and tolerability of oral EPI-506. Secondary objectives included determination of the maximal tolerated dose (MTD), pharmacokinetic profile, and antitumor efficacy. RESULTS 28 mCRPC patients were enrolled into 7 dose cohorts of EPI-506 ranging from 80-3600 mg given once daily and 1800 mg given twice daily. Six DLTs occurred in 4 patients; Grade 4 elevated amylase; Grade 3 abdominal pain; Grade 3 elevated ALT and Grade 3 elevated AST; Grade 2 nausea and Grade 1 vomiting which resulted in study drug intake of
- Published
- 2021
32. Ripretinib intrapatient dose escalation after disease progression provides clinically meaningful outcomes in advanced gastrointestinal stromal tumour
- Author
-
Rodrigo Ruiz-Soto, Filip Janku, Suzanne George, Neeta Somaiah, Robin L. Jones, Albiruni Ryan Abdul Razak, Ying Su, Hans Gelderblom, Michael S. Gordon, Kristen N. Ganjoo, Julia Jennings, Julie Meade, Ping Chi, Jonathan C. Trent, Margaret von Mehren, K. Shi, and Michael Heinrich
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Stromal cell ,Gastrointestinal Stromal Tumors ,medicine.drug_class ,Ripretinib ,Article ,Tyrosine-kinase inhibitor ,Internal medicine ,medicine ,Humans ,Urea ,Dosing ,Progression-free survival ,Naphthyridines ,Adverse effect ,Protein Kinase Inhibitors ,Pharmacology ,Gastrointestinal stromal tumours ,medicine.diagnostic_test ,GiST ,business.industry ,Progression-Free Survival ,Regimen ,Treatment Outcome ,Positron emission tomography ,Disease Progression ,Female ,business - Abstract
Purpose: Ripretinib is a switch-control tyrosine kinase inhibitor that broadly inhibits KIT and platelet-derived growth factor receptor a kinase signalling. Ripretinib showed preliminary efficacy in patients with advanced gastrointestinal stromal tumour (GIST) in a phase I study across a range of doses. Results were confirmed in the phase III INVICTUS study, and ripretinib 150 mg once daily (QD) was subsequently approved as a >fourth-line therapy. Here, we report the phase I study results of intrapatient dose escalation (IPDE) in patients with GIST treated across second, third and later lines of therapy. Methods: Patients with advanced GIST who experienced disease progression (PD) at ripretinib 150 mg QD could dose escalate to 150 mg twice daily (BID). Progression-free survival (PFS) 1 was calculated from the date of the first dose of ripretinib 150 mg QD to PD (as per Response Evaluation Criteria in Solid Tumours 1.1); PFS2 was from the date of IPDE (150 mg BID) to PD or death. Treatment-emergent adverse events (TEAEs) were summarised by dosing periods and compared descriptively. Results: Of 142 patients with GIST receiving ripretinib 150 mg QD, 67 underwent IPDE. IPDE provided benefit across all lines of therapy; the median PFS2 was 5.6, 3.3 and 4.6 months for patients on second-, third-and >fourth-line therapy, respectively. A partial metabolic response after IPDE was demonstrated in 13 of 37 patients with available positron emission tomography scans. TEAEs reported at both doses were similar. Conclusion: Ripretinib IPDE after PD provided continued clinical benefit in advanced GIST across second, third and later lines of therapy with a similar safety profile to that observed with the QD regimen. (c) 2021 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
- Published
- 2021
33. The value of comprehensive genomic sequencing to maximize the identification of clinically actionable alterations in advanced cancer patients: a case series
- Author
-
Laurie J. Goodman, Thomas Royce, Audrey A. Ozols, Gargi D. Basu, Kevin Drenner, Margaux Steinbach, Janine LoBello, Michael S. Gordon, Sunil Sharma, Erkut Borazanci, and Jeffrey M. Trent
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,precision medicine ,Genomic sequencing ,Case Report ,RNA sequencing ,RNA-Seq ,Disease ,rare mutations ,Metastatic tumor ,Precision medicine ,Advanced cancer ,Germline ,whole exome sequencing ,Internal medicine ,Medicine ,business ,genomic alteration ,Exome sequencing - Abstract
Purpose: We present seven cases of advanced cancer patients who initially underwent tumor testing utilizing smaller, panel-based tests, followed by a variety of therapeutic treatments which ultimately resulted in progression of their disease. These cases demonstrate the value of utilizing WES/RNA seq and characterization following disease progression in these patients and the determination of clinically targetable alterations as well as acquired resistance mutations. Materials and Methods: All patients are part of an IRB approved observational study. WES and RNA sequencing were performed, using GEM ExTra® on tumor and blood samples obtained during routine clinical care. To accurately determine somatic versus germline alterations the test was performed with paired normal testing from peripheral blood. Results: The presented cases demonstrate the clinical impact of actionable findings uncovered using GEM ExTra® in patients with advanced disease who failed many rounds of treatment. Unique alterations were identified resulting in newly identified potential targeted therapies, mechanisms of resistance, and variation in the genomic characterization of the primary versus the metastatic tumor. Conclusions: Taken together our results demonstrate that GEM ExTra® maximizes detection of actionable mutations, thus allowing for appropriate treatment selection for patients harboring both common and rare genomic alterations.
- Published
- 2021
34. Phase I Open-Label Study Evaluating the Safety, Pharmacokinetics, and Preliminary Efficacy of Dilpacimab in Patients with Advanced Solid Tumors
- Author
-
Susan E. Morgan-Lappe, Zev A. Wainberg, Ramesh K. Ramanathan, Huibin Yue, Martha Elizabeth Blaney, Louie Naumovski, John H. Strickler, Lan Wang, John Nemunaitis, George W. Sledge, Erika Hamilton, Sreeneeranj Kasichayanula, Minal A. Barve, Michael S. Gordon, and Monica Motwani
- Subjects
Vascular Endothelial Growth Factor A ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Gastroenterology ,Antibodies ,Prostate cancer ,Rare Diseases ,Pharmacokinetics ,Clinical Research ,Gastrointestinal perforation ,Neoplasms ,Internal medicine ,Antibodies, Bispecific ,medicine ,Humans ,Tissue Distribution ,Oncology & Carcinogenesis ,Adverse effect ,Adaptor Proteins, Signal Transducing ,Aged ,Cancer ,business.industry ,Calcium-Binding Proteins ,Signal Transducing ,Adaptor Proteins ,Evaluation of treatments and therapeutic interventions ,Pharmacology and Pharmaceutical Sciences ,Middle Aged ,Prognosis ,medicine.disease ,Ovarian Cancer ,Orphan Drug ,Oncology ,Tolerability ,6.1 Pharmaceuticals ,Pharmacodynamics ,Bispecific ,Female ,Patient Safety ,Ovarian cancer ,business ,Progressive disease ,Follow-Up Studies - Abstract
Dilpacimab (formerly ABT-165), a novel dual-variable domain immunoglobulin, targets both delta-like ligand 4 (DLL4) and VEGF pathways. Here, we present safety, pharmacokinetic (PK), pharmacodynamic (PD), and preliminary efficacy data from a phase I study (trial registration ID: NCT01946074) of dilpacimab in patients with advanced solid tumors. Eligible patients (≥18 years) received dilpacimab intravenously on days 1 and 15 in 28-day cycles at escalating dose levels (range, 1.25–7.5 mg/kg) until progressive disease or unacceptable toxicity. As of August 2018, 55 patients with solid tumors were enrolled in the dilpacimab monotherapy dose-escalation and dose-expansion cohorts. The most common treatment-related adverse events (TRAE) included hypertension (60.0%), headache (30.9%), and fatigue (21.8%). A TRAE of special interest was gastrointestinal perforation, occurring in 2 patients (3.6%; 1 with ovarian and 1 with prostate cancer) and resulting in 1 death. The PK of dilpacimab showed a half-life ranging from 4.9 to 9.5 days, and biomarker analysis demonstrated that the drug bound to both VEGF and DLL4 targets. The recommended phase II dose for dilpacimab monotherapy was established as 3.75 mg/kg, primarily on the basis of tolerability through multiple cycles. A partial response was achieved in 10.9% of patients (including 4 of 16 patients with ovarian cancer). The remaining patients had either stable disease (52.7%), progressive disease (23.6%), or were deemed unevaluable (12.7%). These results demonstrate that dilpacimab monotherapy has an acceptable safety profile, with clinical activity observed in patients with advanced solid tumors.
- Published
- 2021
35. Oral Sabizabulin for High-Risk, Hospitalized Adults with Covid-19: Interim Analysis
- Author
-
K. Gary Barnette, Michael S. Gordon, Domingo Rodriguez, T. Gary Bird, Alan Skolnick, Michael Schnaus, Paula K. Skarda, Suzana Lobo, Eduardo Sprinz, Georgi Arabadzhiev, Petar Kalaydzhiev, and Mitchell Steiner
- Published
- 2022
36. Continuing Care App for Probationers and Parolees with Substance use Disorders
- Author
-
Steven B. Carswell, Michael S. Gordon, Jan Gryczynski, Faye S. Taxman, Mary Schadegg, Kaitlin N. Ferguson, and Kelly Maher
- Subjects
Adult ,Male ,Health (social science) ,Substance-Related Disorders ,Public Health, Environmental and Occupational Health ,Medicine (miscellaneous) ,General Medicine ,Criminals ,Mobile Applications ,Psychiatry and Mental health ,Surveys and Questionnaires ,Outpatients ,Humans ,Female - Abstract
This pilot proof-of-concept study examined the feasibility and acceptability of a Continuing Care mobile application (app) designed to meet the recovery and personal support needs of individuals under justice supervision who were receiving outpatient substance use disorder (SUD) treatment. The study included adults on probation or parole who were enrolled in an outpatient SUD treatment program ( N = 15; 86.7% males). Participants were instructed to utilize the Continuing Care app daily for 4 weeks. At the end of the study, they completed a satisfaction questionnaire. Of the 15 participants enrolled in the study, 12 (80%) completed the Continuing Care app modules and the satisfaction questionnaire, and all of these participants indicated high levels of satisfaction with the app (on a scale of 1–10, Mean = 1.8, SD = 1.2). The Continuing Care app was well-utilized and perceived as valuable by this group of low-income, underserved, and hard-to-reach individuals. Further research is needed to refine app content and evaluate its ability to meaningfully enhance and extend the benefits of SUD treatment.
- Published
- 2022
37. Avelumab Plus Axitinib as First-Line Therapy for Advanced Renal Cell Carcinoma: Long-Term Results from the JAVELIN Renal 100 Phase Ib Trial
- Author
-
James Larkin, Mototsugu Oya, Marcella Martignoni, Fiona Thistlethwaite, Paul Nathan, Moshe C Ornstein, Thomas Powles, Kathryn E Beckermann, Arjun V Balar, David McDermott, Sumati Gupta, George K Philips, Michael S Gordon, Hirotsugu Uemura, Yoshihiko Tomita, Jing Wang, Elisabete Michelon, Alessandra di Pietro, and Toni K Choueiri
- Subjects
Cancer Research ,Oncology - Abstract
Background Progression-free survival was significantly longer in patients who received avelumab plus axitinib versus sunitinib as first-line treatment for advanced renal cell carcinoma (aRCC) in a randomized phase III trial. We report long-term safety and efficacy of avelumab plus axitinib as first-line treatment for patients with aRCC from the JAVELIN Renal 100 phase Ib trial (NCT02493751). Materials and Methods In this open-label, multicenter, phase Ib study, patients with untreated aRCC received avelumab 10 mg/kg every 2 weeks plus axitinib 5 mg twice daily or with axitinib for 7 days followed by avelumab plus axitinib. Safety and efficacy were assessed in all patients receiving at least one dose of avelumab or axitinib. Results Overall, 55 patients were enrolled and treated. Median follow-up was 55.7 months (95% CI, 54.5-58.7). Treatment-related adverse events of any grade or grade ≥3 occurred in 54 (98.2%) and 34 (61.8%) patients, respectively. The confirmed objective response rate was 60.0% (95% CI, 45.9-73.0), including complete response in 10.9% of patients. Median duration of response was 35.9 months (95% CI, 12.7-52.9); the probability of response was 65.8% (95% CI, 46.7-79.4) at 2 years. Median progression-free survival was 8.3 months (95% CI, 5.3-32.0). Median overall survival was not reached (95% CI, 40.8-not estimable); the 5-year overall survival rate was 57.3% (95% CI, 41.2-70.5). Conclusion Five-year follow-up for combination treatment with avelumab plus axitinib in previously untreated patients with aRCC showed long-term clinical activity with no new safety signals, supporting use of this regimen within its approved indication in clinical practice (Clinicaltrials.gov NCT02493751).
- Published
- 2022
38. Safety and Clinical Activity of Atezolizumab in Patients with Metastatic Castration-Resistant Prostate Cancer: A Phase I Study
- Author
-
Yohann Loriot, Indrani Sarkar, Daniel P. Petrylak, Joseph Kim, Paul Conkling, Thomas Powles, Susheela Carroll, Lauren C. Harshman, Jean-Pierre Delord, Edward E. Kadel, Sujata Narayanan, Marcella Fassò, David R. Shaffer, Michael S. Gordon, Sanjeev Mariathasan, Kobe C. Yuen, John D. Powderly, Fadi Braiteh, and Carol O'Hear
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,medicine ,Humans ,Enzalutamide ,In patient ,Adverse effect ,Response Evaluation Criteria in Solid Tumors ,Aged ,business.industry ,Middle Aged ,medicine.disease ,Confidence interval ,Phase i study ,Survival Rate ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,business - Abstract
Purpose: Atezolizumab [anti–programmed death-ligand 1 (anti-PD-L1)] is well tolerated and efficacious in multiple cancers, but has not been previously evaluated in metastatic castration-resistant prostate cancer (mCRPC). This study examined the safety, efficacy, and biomarkers of atezolizumab monotherapy for mCRPC. Patients and Methods: This phase Ia, open-label, dose-escalation and dose-expansion study (PCD4989g) enrolled patients with mCRPC who had progressed on sipuleucel-T or enzalutamide. Atezolizumab was given intravenously every 3 weeks until confirmed disease progression or loss of clinical benefit. Prespecified endpoints included safety, efficacy, biomarker analyses, and radiographic assessments. Results: All 35 evaluable patients [median age, 68 years (range, 45–83 years)] received atezolizumab after ≥1 prior line of therapy; 62.9% of patients had received ≥3 prior lines. Treatment-related adverse events occurred in 21 patients (60.0%), with no deaths. One patient had a confirmed partial response (PR) per RECIST 1.1, and 1 patient had a PR per immune-related response criteria. The confirmed 50% PSA response rate was 8.6% (3 patients). Median overall survival (OS) was 14.7 months [95% confidence interval (CI): 5.9–not evaluable], with a 1-year OS rate of 52.3% (95% CI: 34–70); 2-year OS was 35.9% (95% CI: 13–59). Median follow-up was 13.0 months (range, 1.2–28.1 months). Biomarker analyses showed that atezolizumab activated immune responses; however, a composite biomarker failed to reveal consistent correlations with efficacy. Conclusions: Atezolizumab was generally well tolerated in patients with mCRPC, with a safety profile consistent with other tumor types. In heavily pretreated patients, atezolizumab monotherapy demonstrated evidence of disease control; however, its limited efficacy suggests a combination approach may be needed.
- Published
- 2021
39. Opaganib in Coronavirus Disease 2019 Pneumonia: Results of a Randomized, Placebo-Controlled Phase 2a Trial
- Author
-
Kevin L, Winthrop, Alan W, Skolnick, Adnan M, Rafiq, Scott H, Beegle, Julian, Suszanski, Guenther, Koehne, Ofra, Barnett-Griness, Aida, Bibliowicz, Reza, Fathi, Patricia, Anderson, Gilead, Raday, Gina, Eagle, Vered Katz, Ben-Yair, Harold S, Minkowitz, Mark L, Levitt, and Michael S, Gordon
- Subjects
Infectious Diseases ,Oncology - Abstract
Background Opaganib, an oral sphingosine kinase-2 inhibitor with antiviral and anti-inflammatory properties, was shown to inhibit severe acute respiratory syndrome coronavirus 2 replication in vitro. We thus considered that opaganib could be beneficial for moderate to severe coronavirus disease 2019 (COVID-19) pneumonia. The objective of the study was to evaluate the safety of opaganib and its effect on supplemental oxygen requirements and time to hospital discharge in COVID-19 pneumonia hospitalized patients requiring supplemental oxygen. Methods This Phase 2a, randomized, double-blind, placebo-controlled study was conducted between July and December 2020 in 8 sites in the United States. Forty-two enrolled patients received opaganib (n = 23) or placebo (n = 19) added to standard of care for up to 14 days and were followed up for 28 days after their last dose of opaganib/placebo. Results There were no safety concerns arising in this study. The incidence of ≥Grade 3 treatment-emergent adverse events was 17.4% and 33.3% in the opaganib and placebo groups, respectively. Three deaths occurred in each group. A numerical advantage for opaganib over placebo was observed in in this nonpowered study reflected by total supplemental oxygen requirement from baseline to Day 14, the requirement for supplemental oxygen for at least 24 hours by Day 14, and hospital discharge. Conclusions In this proof-of-concept study, hypoxic, hospitalized patients receiving oral opaganib had a similar safety profile to placebo-treated patients, with preliminary evidence of benefit for opaganib as measured by supplementary oxygen requirement and earlier hospital discharge. These findings support further evaluation of opaganib in this population.
- Published
- 2022
40. Safety and Efficacy of Andecaliximab (GS-5745) Plus Gemcitabine and Nab-Paclitaxel in Patients with Advanced Pancreatic Adenocarcinoma: Results from a Phase I Study
- Author
-
Marianna Zavodovskaya, Jorge Chaves, JieJane Liu, Carrie Baker Brachmann, Jordan Berlin, Michael S. Gordon, Sunil Sharma, Manish R. Patel, Zev A. Wainberg, Pankaj Bhargava, Dung Thai, Kevin S Windsor, Johanna C. Bendell, Saad A. Khan, Manish A. Shah, and Alexander Starodub
- Subjects
0301 basic medicine ,Oncology ,Andecaliximab ,Cancer Research ,medicine.medical_treatment ,Matrix metalloproteinase 9 ,Peripheral edema ,Deoxycytidine ,0302 clinical medicine ,Gastrointestinal Cancer ,Antineoplastic Combined Chemotherapy Protocols ,Monoclonal ,Tumor Microenvironment ,Humanized ,6.2 Cellular and gene therapies ,Cancer ,Treatment Outcome ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Adenocarcinoma ,Patient Safety ,medicine.symptom ,medicine.drug ,medicine.medical_specialty ,Paclitaxel ,Nausea ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antibodies, Monoclonal, Humanized ,Antibodies ,Pancreatic Cancer ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Albumins ,Internal medicine ,medicine ,Carcinoma ,Humans ,GS-5745 ,Oncology & Carcinogenesis ,Adverse effect ,Chemotherapy ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Gemcitabine ,Pancreatic Neoplasms ,Orphan Drug ,030104 developmental biology ,Digestive Diseases ,business ,Pancreatic adenocarcinoma - Abstract
Background Matrix metalloproteinase 9 (MMP9) expression in the tumor microenvironment is implicated in multiple protumorigenic processes. Andecaliximab (GS-5745), a monoclonal antibody targeting MMP9 with high affinity and selectivity, was evaluated in combination with gemcitabine and nab-paclitaxel in patients with advanced pancreatic adenocarcinoma. Patients and Methods This phase I study was completed in two parts: part A was a dose-finding, monotherapy phase that enrolled patients with advanced solid tumors, and part B examined andecaliximab in combination with chemotherapy in specific patient cohorts. In the cohort of patients with pancreatic adenocarcinoma (n = 36), andecaliximab 800 mg every 2 weeks was administered in combination with gemcitabine and nab-paclitaxel. Patients were treated until unacceptable toxicity, withdrawal of consent, disease progression, or death. Efficacy, safety, and biomarker assessments were performed. Results Andecaliximab combined with gemcitabine and nab-paclitaxel appeared to be well tolerated and did not demonstrate any unusual toxicities in patients with pancreatic adenocarcinoma. The most common treatment-emergent adverse events were fatigue (75.0%), alopecia (55.6%), peripheral edema (55.6%), and nausea (50.0%). Median progression-free survival was 7.8 months (90% confidence interval, 6.9−11.0) with an objective response rate of 44.4% and median duration of response of 7.6 months. Maximal andecaliximab target binding, defined as undetectable, andecaliximab-free MMP9 in plasma, was observed. Conclusion Andecaliximab in combination with gemcitabine and nab-paclitaxel demonstrates a favorable safety profile and clinical activity in patients with advanced pancreatic adenocarcinoma. Implications for Practice The combination of andecaliximab, a novel, first-in-class inhibitor of matrix metalloproteinase 9, with gemcitabine and nab-paclitaxel in patients with advanced pancreatic adenocarcinoma provided a median progression-free survival of 7.8 months and objective response rate of 44.4%. The majority of systemic biomarkers related to matrix metalloproteinase 9 activity and immune suppression increased at 2 months, whereas biomarkers related to tumor burden decreased. Although this study demonstrates promising results with andecaliximab plus chemotherapy in patients with advanced pancreatic adenocarcinoma, andecaliximab was not associated with a survival benefit in a phase III study in patients with advanced gastric/gastroesophageal junction carcinoma.
- Published
- 2020
41. Evaluation of Potential Drug‐Drug Interaction Risk of Pexidartinib With Substrates of Cytochrome P450 and P‐Glycoprotein
- Author
-
Michael S. Gordon, Andrew J. Wagner, Fumiaki Kobayashi, Hamim Zahir, Cynthia Zamora, Roohi Gajee, Jonathan Greenberg, Qiang Wang, and Hani M. Babiker
- Subjects
Adult ,Male ,Digoxin ,cytochrome P450 ,CYP3A ,Aminopyridines ,CYP2C19 ,Pharmacology ,030226 pharmacology & pharmacy ,03 medical and health sciences ,0302 clinical medicine ,Tolbutamide ,Pharmacokinetics ,P‐glycoprotein ,medicine ,Cytochrome P-450 CYP3A ,Humans ,Drug Interactions ,Pyrroles ,Pharmacology (medical) ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,CYP2C9 ,Omeprazole ,Aged ,Cytochrome P-450 CYP2C9 ,drug interaction ,Cross-Over Studies ,Chemistry ,Middle Aged ,Drug interaction ,Cytochrome P-450 CYP2C19 ,Area Under Curve ,030220 oncology & carcinogenesis ,Female ,pharmacokinetics ,pexidartinib ,medicine.drug - Abstract
Pexidartinib is approved for treatment of adults with symptomatic tenosynovial giant cell tumor. In vitro data showed pexidartinib's potential to inhibit and induce cytochrome P450 (CYP) 3A, inhibit CYP2C9, CYP2C19 and P‐glycoprotein (P‐gp). Herein, 2 open‐label, single‐sequence, crossover studies evaluated the drug‐drug interaction potential of pexidartinib on CYP enzymes (CYP2C9, CYP2C19, and CYP3A) and P‐gp. Thirty‐two subjects received single oral doses of midazolam (CYP3A substrate) and tolbutamide (CYP2C9 substrate) alone and after single and multiple oral doses of pexidartinib. Twenty subjects received single oral doses of omeprazole (CYP2C19 substrate) and digoxin (P‐gp substrate) alone or with pexidartinib. Analysis of variance was conducted to determine the effect of pexidartinib on various substrates’ pharmacokinetics. No drug‐drug interaction was concluded if the 90% confidence interval of the ratio of test to reference was within the range 80% to 125%. Coadministration of single and multiple doses of pexidartinib resulted in 21% and 52% decreases, respectively, in the area under the plasma concentration–time curve from time zero to the last measurable time point (AUClast) of midazolam, whereas AUClast values of tolbutamide increased 15% and 36%, respectively. Omeprazole exposure decreased on concurrent administration with pexidartinib, the metabolite‐to‐parent ratio was similar following omeprazole administration alone vs coadministration with pexidartinib; pexidartinib did not affect CYP2C19‐mediated metabolism. Maximum plasma concentrations of digoxin slightly increased (32%) with pexidartinib coadministration; no significant effect on digoxin AUClast. These results indicate that pexidartinib is a moderate inducer of CYP3A and a weak inhibitor of CYP2C9 and does not significantly affect CYP2C19‐mediated metabolism or P‐gp transport.
- Published
- 2020
42. Molecular profiling of melanoma brain metastases compared to primary cutaneous melanoma and to extracranial metastases
- Author
-
Jacob Stephen Thomas, Geoffrey T. Gibney, Jeffrey M. Farma, Anthony J. Olszanski, Steven J. O'Day, Steven Daveluy, Michael S. Gordon, Ari M. Vanderwalde, Michael B. Atkins, Lawrence E. Flaherty, Gino K. In, Burton L. Eisenberg, Michelle Saul, Amy Weise, and Kelsey Poorman
- Subjects
PD-L1 ,0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,BRAF ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,SETD2 ,CDKN2A ,brain metastases ,melanoma ,Medicine ,biology ,TMB ,business.industry ,Melanoma ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Cancer research ,biology.protein ,Immunohistochemistry ,business ,Research Paper - Abstract
Background Brain metastases are a significant cause of mortality and morbidity for patients with melanoma. We hypothesize that the development of brain metastases may be explained by molecular heterogeneity between primary cutaneous melanoma (PCM) or extracranial (ECM) and brain (MBM) melanoma metastases. Materials and methods We compared next-generation sequencing, tumor mutational burden (TMB), and immunohistochemical staining for PD-L1 expression, among 132 MBM, 745 PCM, and 1190 ECM. Results The most common genetic alterations among MBM included: BRAF (52.4%), NRAS (26.6%), CDKN2A (23.3%), NF1 (18.9%), TP53 (18%), ARID2 (13.8%), SETD2 (11.9%), and PBRM1 (7.5%). Four genes were found with higher frequency among MBM compared to PCM or ECM: BRAF (52.4% v 40.4% v 40.9%), SETD2 (11.9% v 1.9% v 3.9%), PBRM1 (7.5% v 1.6% v 2.6%), and DICER1 (4.4% v 0.6% v 0.4%). MBM showed higher TMB (p = .04) and higher PD-L1 expression (p = .002), compared to PCM. PD-L1 expression was slightly higher among MBM compared to ECM (p = .042), but there was no difference between TMB (p = .21). Conclusions Our findings suggest a unique molecular profile for MBM, including higher rates of BRAF mutations, higher TMB and higher PD-L1 expression, and also implicate chromatin remodeling in the pathogenesis of MBM.
- Published
- 2020
43. BRAF-Mutant Transcriptional Subtypes Predict Outcome of Combined BRAF, MEK, and EGFR Blockade with Dabrafenib, Trametinib, and Panitumumab in Patients with Colorectal Cancer
- Author
-
Autumn J. McRee, Takayuki Yoshino, Filip de Vos, Chloe E. Atreya, Eduard Gasal, Jan H.M. Schellens, Salvatore Siena, Eric Van Cutsem, Rona Yaeger, Thierry André, John Millholland, Ryan B. Corcoran, Antoine Hollebecque, Catarina D. Campbell, Josep Tabernero, Peter J. O'Dwyer, Fatima Rangwala, Johanna C. Bendell, Jan C. Brase, Yiqun Yang, Gary Middleton, and Michael S. Gordon
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pyridones ,Colorectal cancer ,medicine.medical_treatment ,Oncology and Carcinogenesis ,MAP Kinase Kinase 1 ,Context (language use) ,Pyrimidinones ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Clinical Research ,Internal medicine ,Oximes ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,Humans ,Panitumumab ,Oncology & Carcinogenesis ,Cancer ,Trametinib ,Neoplastic ,Tumor ,business.industry ,Imidazoles ,Dabrafenib ,Cell cycle ,Prognosis ,medicine.disease ,Colo-Rectal Cancer ,Survival Rate ,ErbB Receptors ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Mutation ,Colorectal Neoplasms ,Digestive Diseases ,business ,Biomarkers ,medicine.drug - Abstract
Purpose: The influence of the transcriptional and immunologic context of mutations on therapeutic outcomes with targeted therapy in cancer has not been well defined. BRAF V600E–mutant (BM) colorectal cancer comprises two main transcriptional subtypes, BM1 and BM2. We sought to determine the impact of BM subtype, as well as distinct biological features of those subtypes, on response to BRAF/MEK/EGFR inhibition in patients with colorectal cancer. Patients and Methods: Paired fresh tumor biopsies were acquired at baseline and on day 15 of treatment from all consenting patients with BM colorectal cancer enrolled in a phase II clinical trial of dabrafenib, trametinib, and panitumumab. For each sample, BM subtype, cell cycle, and immune gene signature expression were determined using RNA-sequencing (RNA-seq), and a Cox proportional hazards model was applied to determine association with progression-free survival (PFS). Results: Confirmed response rates, median PFS, and median overall survival (OS) were higher in BM1 subtype patients compared with BM2 subtype patients. Evaluation of immune contexture identified greater immune reactivity in BM1, whereas cell-cycle signatures were more highly expressed in BM2. A multivariate model of PFS incorporating BM subtype plus immune and cell-cycle signatures revealed that BM subtype encompasses the majority of the effect. Conclusions: BM subtype is significantly associated with the outcome of combination dabrafenib, trametinib, and panitumumab therapy and may serve as a standalone predictive biomarker beyond mutational status. Our findings support a more nuanced approach to targeted therapeutic decisions that incorporates assessment of transcriptional context.
- Published
- 2020
44. Phase Ia Study of Anti-NaPi2b Antibody–Drug Conjugate Lifastuzumab Vedotin DNIB0600A in Patients with Non–Small Cell Lung Cancer and Platinum-Resistant Ovarian Cancer
- Author
-
David R. Spigel, Julie Cordova, Michael S. Gordon, Stephanie Royer-Joo, YounJeong Choi, Vanessa Lemahieu, Joan H. Schiller, David E. Gerber, Randall C. Dere, David S. Shames, Maria Martinez Garcia, Anjali Vaze, Enriqueta Felip, Jian Xu, Valerie Westcott, Robert Kahn, Eva Schuth, Sarah B. Goldberg, Daniel J. Maslyar, Katie Wood, Yulei Wang, Jeffrey R. Infante, Eric W. Humke, Kedan Lin, Howard A. Burris, Miguel Martin, and Divya Samineni
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Antibody-drug conjugate ,Immunoconjugates ,Lung Neoplasms ,Maximum Tolerated Dose ,Organoplatinum Compounds ,Nausea ,Carcinoma, Ovarian Epithelial ,Neutropenia ,Antibodies, Monoclonal, Humanized ,Sodium-Phosphate Cotransporter Proteins, Type IIb ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Tissue Distribution ,Lung cancer ,Aged ,Aged, 80 and over ,business.industry ,Middle Aged ,medicine.disease ,Treatment Outcome ,030104 developmental biology ,Oncology ,Monomethyl auristatin E ,chemistry ,Tolerability ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Vomiting ,Female ,Patient Safety ,medicine.symptom ,business - Abstract
Purpose: This phase I trial assessed the safety, tolerability, and preliminary antitumor activity of lifastuzumab vedotin (LIFA), an antibody–drug conjugate of anti-NaPi2b mAb (MNIB2126A) and a potent antimitotic agent (monomethyl auristatin E). Patients and Methods: LIFA was administered to patients with non–small cell lung cancer (NSCLC) and platinum-resistant ovarian cancer (PROC), once every 3 weeks, by intravenous infusion. The starting dose was 0.2 mg/kg in this 3+3 dose-escalation design, followed by cohort expansion at the recommended phase II dose (RP2D). Results: Overall, 87 patients were treated at doses between 0.2 and 2.8 mg/kg. The MTD was not reached; 2.4 mg/kg once every 3 weeks was selected as the RP2D based on overall tolerability profile. The most common adverse events of any grade and regardless of relationship to study drug were fatigue (59%), nausea (49%), decreased appetite (37%), vomiting (32%), and peripheral sensory neuropathy (29%). Most common treatment-related grade ≥3 toxicities among patients treated at the RP2D (n = 63) were neutropenia (10%), anemia (3%), and pneumonia (3%). The pharmacokinetic profile was dose proportional. At active doses ≥1.8 mg/kg, partial responses were observed in four of 51 (8%) patients with NSCLC and 11 of 24 (46%) patients with PROC per RECIST. All RECIST responses occurred in patients with NaPi2b-high by IHC. The CA-125 biomarker assessed for patients with PROC dosed at ≥1.8 mg/kg showed 13 of 24 (54%) had responses (≥50% decline from baseline). Conclusions: LIFA exhibited dose-proportional pharmacokinetics and an acceptable safety profile, with encouraging activity in patients with PROC at the single-agent RP2D of 2.4 mg/kg.
- Published
- 2020
45. A randomized pilot study of Repetitive Transcranial Magnetic Stimulation for Adolescents with Major Depressive Disorder
- Author
-
Michael S. Gordon, Emily J . Wallman, Maxwell Fraser, Christine Pavlou, Paul B Fitzgerald, and Glenn. A Melvin
- Subjects
mental disorders ,behavioral disciplines and activities - Abstract
Objectives: Up to 40% of adolescents with Major Depressive Disorder (MDD) do not respond to treatment. Repetitive Transcranial Magnetic Stimulation (rTMS) is an effective treatment in adults for MDD which shows promising efficacy in adolescents, however additional controlled investigations are needed. This pilot study compared rTMS applied to the left-side of the head and right-side in a randomized controlled trial. We hypothesized rTMS treatment would lead to an improvement in Children’s Depression Rating Scale Revised (CDRS-R) depression scores of adolescents with MDD at 1-month follow-up compared to baseline. No difference in response between left- and right-sided rTMS was expected.Methods: Fourteen adolescents who had been previously treated for MDD but had continued to suffer with MDD for an average of 2 years were randomly allocated to right (low frequency) or left (high frequency) rTMS. rTMS was applied to the scalp over the dorsolateral prefrontal cortex over 20 treatments given over four weeks. The primary outcome measure was the CDRS-R. Outcome assessments occurred at 10 sessions and 20 sessions, with follow-up at one month and six months.Results: CDRS-R scores improved significantly across the 20 rTMS treatments, with peak response at 1-month follow-up. Two (14%) adolescents had ≥ 50% score improvement and a further four (29%) demonstrated partial response (between 25 and 50% reduction) by 1-month follow-up. Treatment gains were sustained at 6-month follow-up. There was no significant difference in efficacy between left- and right-sided treatment.Conclusion: This small study found preliminary evidence that adolescents with MDD benefited from rTMS.
- Published
- 2022
46. Phase Ib Study of the Histone Deacetylase 6 Inhibitor Citarinostat in Combination With Paclitaxel in Patients With Advanced Solid Tumors
- Author
-
Michael S. Gordon, Geoffrey I. Shapiro, John Sarantopoulos, Dejan Juric, Brian Lu, Angeliki Zarotiadou, Jamie N. Connarn, Yvan Le Bruchec, Calin Dan Dumitru, and R. Donald Harvey
- Subjects
paclitaxel ,Cancer Research ,epigenetics ,Oncology ,histone deacetylase ,histone acetylation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,advanced solid tumors ,HDAC inhibition ,citarinostat ,RC254-282 ,Original Research ,combination therapy - Abstract
BackgroundCitarinostat (CC-96241; previously ACY-241), an oral inhibitor of histone deacetylases (HDACs) with selectivity for HDAC6, has demonstrated synergistic anticancer activity with paclitaxel in multiple solid tumor models. Combination therapy using citarinostat with paclitaxel was evaluated in this phase Ib 3 + 3 dose-escalation study in patients with advanced solid tumors.MethodsPatients with previously treated advanced solid tumors received citarinostat 180, 360, or 480 mg once daily on days 1 to 21 plus paclitaxel 80 mg/m2 on days 1, 8, and 15 of 28-day cycles until disease progression or unacceptable toxicity. The primary endpoint was determination of the maximum tolerated dose (MTD). Secondary endpoints included safety, antitumor activity, pharmacokinetics, and pharmacodynamics.ResultsTwenty patients were enrolled and received study treatment; 15 had received prior taxane therapy. No dose-limiting toxicities were reported at any dose; therefore, the MTD was not identified. Citarinostat 360 vs 480 mg was associated with reduced incidence and severity of neutropenia. Three patients experienced a confirmed partial response and 13 achieved stable disease. Pharmacokinetic parameters were linear up to citarinostat 360 mg, the dose at which the highest levels of histone and tubulin acetylation were observed in peripheral blood mononuclear cells.ConclusionsThe combination of citarinostat plus paclitaxel showed an acceptable safety profile, with no unexpected or dose-limiting toxicities and potential evidence of antitumor activity in patients with heavily pretreated advanced solid tumors. Citarinostat 360 mg once daily is considered the recommended phase II dose for use in combination with paclitaxel 80 mg/m2 every 3 of 4 weeks. This trial is registered on ClinicalTrials.gov (NCT02551185).
- Published
- 2022
47. Phase 1 efficacy and pharmacodynamic results of exicorilant + enzalutamide in patients with metastatic castration-resistant prostate cancer
- Author
-
Michael J. Morris, Mark David Linch, Simon J. Crabb, Tomasz M. Beer, Elisabeth I. Heath, Michael S. Gordon, Johann S. De Bono, Hristina I Pashova, Iulia Cristina Tudor, Andrew E Greenstein, Grace Mann, and Glenn Liu
- Subjects
Cancer Research ,Oncology - Abstract
145 Background: Metastatic castration-resistant prostate cancer (mCRPC) remains an incurable disease with significant morbidity. Androgen receptor (AR) signaling is a key driver of tumor growth in mCRPC, and AR-targeted therapies are the mainstay for patients (pts) with locally advanced or metastatic disease. Enzalutamide (ENZA) is commonly used, but resistance typically develops within 1–2 years. The glucocorticoid receptor (GR) can substitute for the AR, providing a tumor escape pathway (Arora et al. Cell 2013). In the 22Rv1 CRPC xenograft model, the selective GR modulator exicorilant (EXI) combined with ENZA reduced tumor growth, supporting the hypothesis that dual antagonism of GR + AR may block this escape pathway. Safety and pharmacokinetics from the first study of EXI + ENZA in pts with mCRPC (NCT03437941) were previously presented (Linch et al. ESMO 2022). Here, we report efficacy and pharmacodynamic (PD) results from the same study. Methods: Segment 1 (Seg 1) of this phase 1 study evaluated open-label, fasting, BID dosing of EXI (140 or 180 mg) + ENZA 160 mg QD. Segment 2 (Seg 2) tested QD dosing of EXI with food in a double-blind design: All pts received EXI 240 mg + ENZA and were randomized 3:1 to EXI titration (to 280 mg followed by 320 mg) or to remain on EXI 240 mg + placebo. Efficacy assessments included radiographic response and changes in prostate-specific antigen (PSA) levels. PSA was collected prior to study entry and PSA doubling times were calculated before and during treatment. PD analyses included baseline (BL) tumor GR expression and modulation of GR target genes in whole blood. As not all pts enrolled in Seg 1 were ENZA naïve, efficacy data are reported for Seg 2 only. Data cutoff date: July 7, 2022. Results: 39 pts were enrolled (Seg 1: 14, irrespective of prior ENZA exposure; Seg 2: 25, on a stable ENZA dose with rising PSA, defined as a 25% increase over nadir and absolute value >1 ng/mL). In Seg 2, there were no radiographic responses, 18 pts had a best overall response of stable disease per PCWG3, and 1 pt achieved a PSA response (≥50% PSA reduction from BL). BL tumor GR expression was detectable in all assessed tumors. PD analyses demonstrated EXI modulation of GR target genes, such as CDKN1C. Comparable PD effects were observed across EXI doses (240–320 mg QD). While BL 24-h urinary free cortisol (UFC) for most pts was within the normal range, improvements in PSA doubling times after treatment with EXI + ENZA were predominantly observed in pts with higher BL UFC ( P
- Published
- 2023
48. Results from a phase 1a/1b study of botensilimab (BOT), a novel innate/adaptive immune activator, plus balstilimab (BAL; anti-PD-1 antibody) in metastatic heavily pretreated microsatellite stable colorectal cancer (MSS CRC)
- Author
-
Anthony B. El-Khoueiry, Marwan Fakih, Michael S. Gordon, Apostolia Maria Tsimberidou, Andrea J. Bullock, Breelyn A. Wilky, Jonathan C. Trent, Kim Allyson Margolin, Daruka Mahadevan, Ani Sarkis Balmanoukian, Rachel E. Sanborn, Gary K. Schwartz, Bruno Bockorny, Justin C Moser, Joseph Elan Grossman, Waldo Ignacio Ortuzar Feliu, Katherine Rosenthal, Steven O'Day, Heinz-Josef Lenz, and Benjamin L. Schlechter
- Subjects
Cancer Research ,Oncology - Abstract
LBA8 Background: BOT promotes optimized T cell priming, activation and memory formation by strengthening antigen presenting cell/T cell co-engagement. As an Fc-enhanced next-generation anti–CTLA-4 antibody, BOT also promotes intratumoral regulatory T cell depletion and reduces complement fixation. We present results from patients with MSS CRC treated with BOT + BAL in an expanded phase 1a/1b study; NCT03860272. Methods: Patients (pts) with metastatic MSS CRC received BOT 1 or 2 mg/kg every 6 weeks (Q6W) + BAL 3 mg/kg every 2 weeks. Crossover from monotherapy to combination therapy was permitted (rescue) as well as fixed-dosing (150 mg BOT Q6W + 450 mg BAL every 3 weeks). Results: Fifty-nine combination pts were evaluable for efficacy/safety (treated as of 19 May 2022 with ≥1 Q6W imaging assessment), including one rescue and one fixed-dose pt. Median pt age was 57 (range, 25-83), 58% were female, and 76% received at least three prior lines of therapy including prior immunotherapy (34%). Median follow-up was 6.4 months (range, 1.6-29.5). In all pts, objective response rate (ORR) was 22% (95% CI, 12-35), disease control rate (DCR) was 73% (95% CI, 60-84), and median duration of response (DOR) was not reached (NR), with 9/13 responses ongoing. The 12-month overall survival (OS) rate was 61% (95% CI, 42-75), with median OS NR. Of the 13 responders, 9 had RAS mutations (7 KRAS, 2 NRAS), 0 had BRAF mutations, 0/10 had a TMB of ≥10 mutations/Mb, and 1/7 was PD-L1 positive (≥1% combined positive score). A subgroup analysis was conducted by the dose of BOT received . In 1 mg/kg pts (n=8), ORR was 38% (3/8; 95% CI, 9-76) and DCR was 100% (8/8; 95% CI, 63-100); in 2 mg/kg pts (n=50), ORR was 20% (10/50; 95% CI, 10-34) and DCR was 70% (35/50; 95% CI, 55-82). All grade treatment-related adverse events (TRAEs) occurred in 88% of pts, including grade 3 in 32%, and grade 4 in 2% of pts. Diarrhea/colitis was the only grade 3/4 TRAE occurring in more than three pts (15% grade 3, 2% grade 4). The most common grade 3 TRAEs outside of diarrhea/colitis were fatigue (5%) and pyrexia (5%). There were no grade 5 TRAEs reported. Fifteen percent of pts had a TRAE leading to discontinuation of BOT alone and 12% had a TRAE leading to discontinuation of both BOT + BAL. Conclusions: In heavily pretreated metastatic MSS CRC pts, BOT + BAL continues to demonstrate promising clinical activity with durable responses and was well tolerated with no new immune-mediated safety signals. A larger pt set, analyses by subgroup, and additional translational data will be presented at the meeting. A randomized phase 2 trial in MSS CRC pts is enrolling (NCT05608044). Clinical trial information: NCT03860272 .
- Published
- 2023
49. The Unexpected Spectrum of the Innermost Ejecta of the Red Hypergiant VY CMa∗
- Author
-
Roberta M. Humphreys, L. M. Ziurys, J. J. Bernal, Michael S. Gordon, L. Andrew Helton, Kazunori Ishibashi, Terry J. Jones, A. M. S. Richards, and Wouter Vlemmings
- Published
- 2019
- Full Text
- View/download PDF
50. 377 Characterization of peripheral biomarkers of GS-1423, a first in class bifunctional anti-CD73-TGFβ receptor II- trap molecule, in a phase 1 dose escalation study in patients with advanced solid tumors
- Author
-
Kathleen M. Mahoney, Audrey Goddard, Matthew Peach, Marianna Zavodovskaya, Michael S. Gordon, Rick Sorensen, James Strauss, Kai-Wen Lin, Xiaoyun Yang, Tianling Chen, Juliane Jürgensmeier, Anna Seto, Anthony W. Tolcher, Ping Cheng Yi, and Biao Li
- Subjects
Pharmacology ,Cancer Research ,biology ,business.industry ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,Tolerability ,Pharmacokinetics ,biology.protein ,Molecular Medicine ,Immunology and Allergy ,Biomarker (medicine) ,Medicine ,Cytotoxic T cell ,Antibody ,Receptor ,business ,RC254-282 ,Whole blood ,Platelet-poor plasma - Abstract
BackgroundGS-1423 is a first-in-class bifunctional molecule comprised of an anti-CD73 antibody fused to the extracellular domain of TGFβ receptor II (TGFβRII). GS-1423 is designed to inhibit CD73-mediated adenosine production and neutralize active TGFβ within the tumor microenvironment. Dual antagonism of these 2 broadly immunosuppressive barriers is anticipated to facilitate productive anti-tumor immunity.MethodsThis open label Phase 1a study (NCT03954704) evaluated the safety, tolerability, and pharmacokinetics of GS-1423. Exploratory biomarkers included the evaluation of the inhibition of GS-1423 targets, i.e.CD73 and TGFβ, in the periphery. Biomarker assessments were performed in serial blood samples from patients receiving GS-1423 every two weeks (Q2W). Biomarker assays, unless otherwise stated, were custom built and qualified to measure the following: 1) TGF-beta 1/2/3 (Luminex, Bio-Rad) in platelet poor plasma, 2) CD73 target occupancy (TO) on B and CD8 T cells in whole blood, 3) free soluble CD73 (sCD73) not bound to GS-1423, and 4) sCD73 activity in platelet poor plasma. Biomarker values were plotted longitudinally by patient and by dose.ResultsA dose dependent decrease in TGF-beta 1/2/3 in plasma of patients was observed on treatment. There was no detectable TGFβ at the 20 mg/kg dose level and above at 2 hours post first dose and for the duration of the Q2W dosing interval. A dose dependent increase in CD73 TO on B and CD8 T cells was also observed with treatment, and complete TO was achieved at 20 mg/kg and above at 2 hours post first dose for the duration of the Q2W dosing interval. Free sCD73 decreased at 2 hours post first dose, while remaining above the lower limit of quantitation, and then increased above baseline after 24 hours post-dose at the 3 mg/kg dose level and above. The sCD73 activity in blood correlated with changes in free sCD73 levels.ConclusionsBlood biomarker analyses of GS-1423 in patients with advanced solid tumors demonstrated undetectable soluble TGFβ1/2/3 and complete TO of CD73 on B and T cells at the 20 mg/kg dose level and above. The mechanism underlying the increase in sCD73 following GS-1423 treatment remains to be elucidated.Ethics ApprovalThe study obtained ethics approval from the IRB/IEC and all participants gave informed consent before taking part in the study.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.