325 results on '"Donald A. Richards"'
Search Results
2. Supplementary Table 2 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
Grade 3 or 4 adverse events possibly related to refametinib or sorafenib, occurring in {greater than or equal to}2 patients in either cohort
- Published
- 2023
3. Supplementary Table S1. from Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
José Baselga, Jerry Y. Hsu, Michael C. Wei, Thomas J. Stout, Timothy R. Wilson, Na Cui, Alison K. Cardenas, Mafalda Oliveira, Lajos Pusztai, Manish R. Patel, Philippe L. Bedard, Andrés Cervantes, Ian E. Krop, Donald A. Richards, Cristina Saura, and Maura N. Dickler
- Abstract
Study disposition
- Published
- 2023
4. Supplementary Table 3 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
Overview of dose-limiting toxicities and adverse events leading to discontinuation and relationship to study treatment
- Published
- 2023
5. Supplementary Table 5 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
Refametinib geometric mean (% coefficient of variation) multiple-dose pharmacokinetic data on day 1 of course 2
- Published
- 2023
6. Supplementary Figure S1. from Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
José Baselga, Jerry Y. Hsu, Michael C. Wei, Thomas J. Stout, Timothy R. Wilson, Na Cui, Alison K. Cardenas, Mafalda Oliveira, Lajos Pusztai, Manish R. Patel, Philippe L. Bedard, Andrés Cervantes, Ian E. Krop, Donald A. Richards, Cristina Saura, and Maura N. Dickler
- Abstract
Participant PIK3CA-mutation central testing results
- Published
- 2023
7. Supplementary Table 1 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
Incidence of treatment-emergent adverse events, all grades, occurring in {greater than or equal to}20% of patients in either cohort
- Published
- 2023
8. Supplementary Figure 2 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
CTC enumeration and pERK analysis in course 1
- Published
- 2023
9. Supplementary Figure 1 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
pERK levels in hair follicles by dose level and course
- Published
- 2023
10. Supplementary Table 4 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
Refametinib geometric mean (% coefficient of variation) single-dose pharmacokinetic data on day 1 of course 1
- Published
- 2023
11. Data from Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
José Baselga, Jerry Y. Hsu, Michael C. Wei, Thomas J. Stout, Timothy R. Wilson, Na Cui, Alison K. Cardenas, Mafalda Oliveira, Lajos Pusztai, Manish R. Patel, Philippe L. Bedard, Andrés Cervantes, Ian E. Krop, Donald A. Richards, Cristina Saura, and Maura N. Dickler
- Abstract
Purpose: This single-arm, open-label phase II study evaluated the safety and efficacy of taselisib (GDC-0032) plus fulvestrant in postmenopausal women with locally advanced or metastatic HER2-negative, hormone receptor (HR)-positive breast cancer.Patients and Methods: Patients received 6-mg oral taselisib capsules daily plus intramuscular fulvestrant (500 mg) until disease progression or unacceptable toxicity. Tumor tissue (if available) was centrally evaluated for PIK3CA mutations. Adverse events (AE) were recorded using NCI-CTCAE v4.0. Tumor response was investigator-determined using RECIST v1.1.Results: Median treatment duration was 4.6 (range: 0.9–40.5) months. All patients experienced ≥1 AE, 30 (50.0%) had grade ≥3 AEs, and 19 (31.7%) experienced 35 serious AEs. Forty-seven of 60 patients had evaluable tissue for central PIK3CA mutation testing [20 had mutations, 27 had no mutation detected (MND)]. In patients with baseline measurable disease, clinical activity was observed in tumors with PIK3CA mutations [best confirmed response rate: 38.5% (5/13; 95% CI, 13.9–68.4); clinical benefit rate (CBR): 38.5% (5/13; 95% CI, 13.9–68.4)], PIK3CA-MND [best confirmed response rate: 14.3% (3/21; 95% CI, 3.0–36.3); CBR: 23.8% (5/21; 95% CI, 8.2–47.2)], and unknown PIK3CA mutation status [best confirmed response rate: 20.0% (2/10; 95% CI, 2.5–55.6); CBR: 30.0% (3/10; 95% CI, 6.7–65.2)].Conclusions: Taselisib plus fulvestrant had clinical activity irrespective of PIK3CA mutation status, with numerically higher objective response rate and CBR in patients with PIK3CA-mutated (vs. -MND) locally advanced or metastatic HER2-negative, HR-positive breast cancer. No new safety signals were reported. A confirmatory phase III trial is ongoing. Clin Cancer Res; 24(18); 4380–7. ©2018 AACR.
- Published
- 2023
12. Supplementary Table 6 from A Phase I Study of the Safety, Pharmacokinetics, and Pharmacodynamics of Combination Therapy with Refametinib plus Sorafenib in Patients with Advanced Cancer
- Author
-
Neil J. Clendeninn, Heiko Krissel, Prabhu Rajagopalan, Kimberly J. Manhard, David M. Wilson, Sonny Gunawan, Li-Tain Yeh, Zancong Shen, Jeffrey N. Miner, Cory Iverson, Diane P. Leffingwell, Lawrence Garbo, Morris Sherman, Aram F. Hezel, Joe J. Stephenson, Carlos H.R. Becerra, Fadi Braiteh, Anthony El-Khoueiry, Donald A. Richards, and Alex A. Adjei
- Abstract
Sorafenib geometric mean (% coefficient of variation) multiple-dose pharmacokinetic data on day 1 of course 2
- Published
- 2023
13. Data from Trilaciclib Prior to Chemotherapy in Patients with Metastatic Triple-Negative Breast Cancer: Final Efficacy and Subgroup Analysis from a Randomized Phase II Study
- Author
-
Joyce O'Shaughnessy, Janet Horton, Jessica Sorrentino, Jie Xiao, Dušan Milenković, Sharon T. Wilks, Donald A. Richards, Ling Ma, Nikola Vasev, Željko Vojnović, Hyo S. Han, Timothy J. Pluard, Lazar Popovic, Michael A. Danso, Anu R. Thummala, Gail S. Wright, and Antoinette R. Tan
- Abstract
Purpose:We report final antitumor efficacy results from a phase II study of trilaciclib, an intravenous cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, administered prior to gemcitabine plus carboplatin (GCb) in patients with metastatic triple-negative breast cancer (NCT02978716).Patients and Methods:Patients were randomized (1:1:1) to group 1 [GCb (days 1, 8); n = 34], group 2 [trilaciclib prior to GCb (days 1, 8); n = 33], or group 3 [trilaciclib (days 1, 8) and trilaciclib prior to GCb (days 2, 9); n = 35]. Subgroup analyses were performed according to CDK4/6 dependence, level of programmed death-ligand 1 (PD-L1) expression, and RNA-based immune signatures using proportional hazards regression. T-cell receptor (TCR) β CDR3 regions were amplified and sequenced to identify, quantify, and compare the abundance of each unique TCRβ CDR3 at baseline and on treatment.Results:Median overall survival (OS) was 12.6 months in group 1, not reached in group 2 (HR = 0.31; P = 0.0016), 17.8 months in group 3 (HR = 0.40; P = 0.0004), and 19.8 months in groups 2 and 3 combined (HR = 0.37; P < 0.0001). Efficacy outcomes were comparable regardless of cancer CDK4/6 dependence status and immune signatures. Administering trilaciclib prior to GCb prolonged OS irrespective of PD-L1 status but had greater benefit in the PD-L1–positive population. T-cell activation was enhanced in patients receiving trilaciclib.Conclusions:Administering trilaciclib prior to GCb enhanced antitumor efficacy, with significant improvements in OS. Efficacy outcomes in immunologic subgroups and enhancements in T-cell activation suggest these improvements may be mediated via immunologic mechanisms.
- Published
- 2023
14. Supplemental Methods from Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
José Baselga, Jerry Y. Hsu, Michael C. Wei, Thomas J. Stout, Timothy R. Wilson, Na Cui, Alison K. Cardenas, Mafalda Oliveira, Lajos Pusztai, Manish R. Patel, Philippe L. Bedard, Andrés Cervantes, Ian E. Krop, Donald A. Richards, Cristina Saura, and Maura N. Dickler
- Abstract
Definition of adverse events of special interest (AESI)
- Published
- 2023
15. MRTX-500 Phase 2 Trial: Sitravatinib With Nivolumab in Patients With Nonsquamous NSCLC Progressing On or After Checkpoint Inhibitor Therapy or Chemotherapy
- Author
-
Kai He, David Berz, Shirish M. Gadgeel, Wade T. Iams, Debora S. Bruno, Collin M. Blakely, Alexander I. Spira, Manish R. Patel, David M. Waterhouse, Donald A. Richards, Anthony Pham, Robert Jotte, David S. Hong, Edward B. Garon, Anne Traynor, Peter Olson, Lisa Latven, Xiaohong Yan, Ronald Shazer, and Ticiana A. Leal
- Subjects
Pulmonary and Respiratory Medicine ,Oncology - Published
- 2023
16. Value orientations and support for guaranteed income
- Author
-
Thomas L. Steiger and Donald R. Richards
- Subjects
Statistics ,Economics ,General Social Sciences ,Value (mathematics) - Published
- 2021
17. Lenvatinib plus pembrolizumab in patients with either treatment-naive or previously treated metastatic renal cell carcinoma (Study 111/KEYNOTE-146): a phase 1b/2 study
- Author
-
David R. Shaffer, Matthew H. Taylor, Alvaro Pinto, Daniel Heinrich, Chung-Han Lee, Chinyere E. Okpara, Øyvind Krohn Tennøe, Mehmet Asim Bilen, Donald A. Richards, Jane Wu, Randy F. Sweis, Arpit Rao, Rodolfo F. Perini, Amishi Yogesh Shah, Allen Lee Cohn, Jay Courtright, James J. Hsieh, Emmett V. Schmidt, Sara Gunnestad Ribe, Alan D. Smith, Drew W. Rasco, Robert J. Motzer, Regina Gironés Sarrió, Christopher Di Simone, Sharad Jain, Musaberk Goksel, Peter Kubiak, and Nicholas J. Vogelzang
- Subjects
education.field_of_study ,medicine.medical_specialty ,business.industry ,Population ,Pembrolizumab ,Sudden death ,Clinical trial ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Oncology ,chemistry ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Internal medicine ,Clinical endpoint ,medicine ,TRIAL ,030212 general & internal medicine ,COMBINATION ,education ,Lenvatinib ,Adverse effect ,business - Abstract
Summary Background Despite advances in the first-line treatment of metastatic renal cell carcinoma (RCC), there is an unmet need for options to address disease progression during or after treatment with immune checkpoint inhibitors (ICIs). Pembrolizumab and lenvatinib are active as monotherapies in RCC; thus, we aimed to evaluate the combination of lenvatinib plus pembrolizumab in these patients. Methods We report results of the metastatic RCC cohort from an open-label phase 1b/2 study of lenvatinib plus pembrolizumab in patients aged at least 18 years with selected solid tumours and an Eastern Cooperative Oncology Group performance status of 0–1. Oral lenvatinib at 20 mg was given once daily along with intravenous pembrolizumab at 200 mg once every 3 weeks. Patients remained on study drug treatment until disease progression, development of unacceptable toxicity, or withdrawal of consent. Efficacy was analysed in patients with clear cell metastatic RCC receiving study drug by previous therapy grouping: treatment naive, previously treated ICI naive (previously treated with at least one line of therapy but not with an anti-PD-1 or anti-PD-L1 ICI), and ICI pretreated (ie, anti-PD-1 or anti-PD-L1) patients. Safety was analysed in all enrolled and treated patients. The primary endpoint was the objective response rate at week 24 per immune-related Response Evaluation Criteria In Solid Tumors (irRECIST) by investigator assessment. This trial is registered with ClinicalTrials.gov ( NCT02501096 ) and with the EU Clinical Trials Register (EudraCT2017-000300-26), and is closed to new participants. Findings Between July 21, 2015, and Oct 16, 2019, 145 patients were enrolled in the study. Two patients had non-clear cell RCC and were excluded from the efficacy analysis (one in the treatment-naive group and one in the ICI-pretreated group); thus, the population evaluated for efficacy comprised 143 patients (n=22 in the treatment-naive group, n=17 in the previously treated ICI-naive group, and n=104 in the ICI-pretreated group). All 145 enrolled patients were included in the safety analysis. The median follow-up was 19·8 months (IQR 14·3–28·4). The number of patients with an objective response at week 24 by irRECIST was 16 (72·7%, 95% CI 49·8–89·3) of 22 treatment-naive patients, seven (41·2%, 18·4–67·1) of 17 previously treated ICI-naive patients, and 58 (55·8%, 45·7–65·5) of 104 ICI-pretreated patients. Of 145 patients, 82 (57%) had grade 3 treatment-related adverse events and ten (7%) had grade 4 treatment-related adverse events. The most common grade 3 treatment-related adverse event was hypertension (30 [21%] of 145 patients). Treatment-related serious adverse events occurred in 36 (25%) patients, and there were three treatment-related deaths (upper gastrointestinal haemorrhage, sudden death, and pneumonia). Interpretation Lenvatinib plus pembrolizumab showed encouraging antitumour activity and a manageable safety profile and might be an option for post-ICI treatment of metastatic RCC. Funding Eisai and Merck Sharp & Dohme.
- Published
- 2021
18. Phase I Study of Elacestrant (RAD1901), a Novel Selective Estrogen Receptor Degrader, in ER-Positive, HER2-Negative Advanced Breast Cancer
- Author
-
Maureen G. Conlan, Wael A. Harb, Robert Wesolowski, Meghan Sri Karuturi, Donald A. Richards, Peter Kabos, Rebecca G. Bagley, Paul Conkling, Cynthia Osborne, Aditya Bardia, Amy Weise, Sharon Wilks, Virginia G. Kaklamani, and Yamei Wang
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,Tetrahydronaphthalenes ,Receptor, ErbB-2 ,medicine.drug_class ,Advanced breast ,Estrogen receptor ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Receptor ,Aged ,Aged, 80 and over ,business.industry ,Estrogen Receptor alpha ,HER2 negative ,Cancer ,Middle Aged ,medicine.disease ,Phase i study ,030104 developmental biology ,Receptors, Estrogen ,Oncology ,Multicenter study ,Estrogen ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,business - Abstract
PURPOSE This phase I study (RAD1901-005; NCT02338349) evaluated elacestrant, an investigational oral selective estrogen receptor degrader (SERD), in heavily pretreated women with estrogen receptor–positive, human epidermal growth factor receptor 2–negative metastatic breast cancer, including those with estrogen receptor gene alpha ( ESR1) mutation. The primary objective was to determine the maximum tolerated dose and/or recommended phase II dose (RP2D). METHODS The study consisted of a 3 + 3 design (elacestrant capsules) followed by expansion at RP2D (400-mg capsules, then 400-mg tablets) for the evaluation of safety and antitumor activity. Elacestrant was taken once daily until progression or intolerability. RESULTS Of 57 postmenopausal women enrolled, 50 received RP2D (400 mg once daily): median age, 63 years; median three prior anticancer therapies, including cyclin-dependent kinase 4,6 inhibitors (CDK4/6i; 52%), SERD (52%), and ESR1 mutation (circulating tumor DNA; 50%). No dose-limiting toxicities occurred; the most common adverse events at RP2D (400-mg tablet; n = 24) were nausea (33.3%) and increased blood triglycerides and decreased blood phosphorus (25.0% each). Most adverse events were grade 1-2 in severity. The objective response rate was 19.4% (n = 31 evaluable patients receiving RP2D), 15.0% in patients with prior SERD, 16.7% in patients with prior CDK4/6i, and 33.3% in patients with ESR1 mutation (n = 5/15). The clinical benefit rate (24-week) was 42.6% overall (n = 47 patients receiving RP2D), 56.5% (n = 23, ESR1 mutation), and 30.4% (n = 23, prior CDK4/6i). Elacestrant clinical benefit was associated with decline in ESR1 mutant allele fraction. CONCLUSION Elacestrant 400 mg orally once daily has an acceptable safety profile and demonstrated single-agent activity with confirmed partial responses in heavily pretreated patients with estrogen receptor–positive metastatic breast cancer. Notably, responses were observed in patients with ESR1 mutation as well as those with prior CDK4/6i and prior SERD. A phase III trial investigating elacestrant versus standard endocrine therapy is ongoing.
- Published
- 2021
19. Abstract PS11-26: A phase 1 study of D-0502, an orally bioavailable SERD, for advanced or metastatic HR-positive and HER2-negative breast cancer
- Author
-
Donald A. Richards, Amardeep Aulakh, Yanxia Shi, Erika Hamilton, Tao Sun, Sharon Wilks, Kate Lathrop, Andrea Silber, Quchang Ouyang, Sami Diab, Kathryn Stazzone, Ben Cho, Zhe Shi, Cynthia Osborne, Ling Zhang, Binghe Xu, Yaolin Wang, Dejan Juric, and William Jeffery Edenfield
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Fulvestrant ,business.industry ,Estrogen receptor ,Cancer ,Palbociclib ,medicine.disease ,Breast cancer ,Pharmacokinetics ,Tolerability ,Internal medicine ,medicine ,Stage (cooking) ,business ,medicine.drug - Abstract
Background: Targeting the estrogen receptor (ER) has proven to be one of the most successful strategies in treating HR+ (ER+ and PR+) and HER2- breast cancer. Selective estrogen receptor degraders (SERD) can be used as single agents or in combination with CDK4/6 inhibitors such as palbociclib. Fulvestrant is currently the only approved agent in its class and is limited by poor oral bioavailability necessitating monthly intramuscular injections. D-0502 is an orally bioavailable SERD with potent activity in various HR+ and HER2- breast cancer cell lines and xenograft models. Its combination with palbociclib in both MCF-7 xenograft models and ESR-1 mutated (Y537S) patient derived breast cancer xenograft models resulted in further tumor growth inhibition or regression. Drug metabolism and pharmacokinetic studies both in vitro and in vivo demonstrated that D-0502 exhibits favorable PK profiles suitable for clinical development. Methods: A first-in-human phase 1 study was conducted to evaluate D-0502 in women with advanced or metastatic HR+, HER2- breast cancer (MBC) (NCT03471663). The primary objective is to characterize the safety and tolerability of D-0502 and D-0502 in combination with palbociclib, to identify an MTD and/or RP2D. The secondary objectives are to evaluate the PK properties and the preliminary anti-tumor activities. Patients received D-0502 orally once daily in 28-day cycles. The study has completed dose escalation (phase 1a), and dose expansion and combination studies (phase 1b) are ongoing. In phase Ia, patients were enrolled and evaluated using conventional 3+3 dose-escalation to identify the MTD of D-0502 as a single agent. The phase 1b was divided into 2 stages. In Stage 1, D-0502 was evaluated with palbociclib at a dose below the MTD first before escalating to the MTD. Stage 1 also included patients in China treated at a dose below and at the MTD as single agent as well as in combination with palbociclib. Stage 2 will further evaluate the MTD for both single agent and combination of D-0502 with palbociclib. Results: At the time of this abstract submission, phase 1a dose escalation is completed, R2PD has been identified, and no DLTs were observed. PK analysis of D-0502 indicates a dose proportional increase in exposure as the dose increases, and the exposure has exceeded the potential therapeutic exposure level based on preclinical studies. The Phase Ib Stage 1 portion has also completed enrollment and the study is currently enrolling patients into phase Ib Stage 2. D-0502 as a single agent or in combination has been well tolerated with radiological tumor response and clinical benefit response (CBR) observed. Safety, PK and preliminary efficacy data will be reported at the meeting presentation. Conclusion: A first-in-human phase 1 study of D-0502 has been initiated in women with advanced or metastatic HR-positive and HER2-negative breast cancer. D-0502 has been well tolerated and achieved significant exposure and preliminary clinical activity in patients. Further results will be presented at the meeting. Citation Format: Cynthia Osborne, Donald A Richards, Sharon T Wilks, Sami Diab, Dejan Juric, Kate Lathrop, Andrea Silber, William Edenfield, Amardeep Aulakh, Ben Cho, Binghe Xu, Tao Sun, Quchang Ouyang, Yanxia Shi, Kathryn Stazzone, Zhe Shi, Ling Zhang, Yaolin Wang, Erika P Hamilton. A phase 1 study of D-0502, an orally bioavailable SERD, for advanced or metastatic HR-positive and HER2-negative breast cancer [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS11-26.
- Published
- 2021
20. Abstract PD1-06: Trilaciclib improves overall survival when given with gemcitabine/carboplatin in patients with metastatic triple-negative breast cancer: Final analysis of a randomized phase 2 trial
- Author
-
Lazar Popovic, Hyo S. Han, Jessica A. Sorrentino, Gail S. Wright, Jie Xiao, Željko Vojnović, Anu Thummala, Donald A. Richards, Sharon Wilks, Dušan Milenković, Michael A. Danso, Nikola Vasev, Joyce O'Shaughnessy, Antoinette R. Tan, Janet K. Horton, Ling Ma, and Timothy J. Pluard
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Gemcitabine ,Carboplatin ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,Overall survival ,In patient ,business ,Triple-negative breast cancer ,medicine.drug - Abstract
Background Trilaciclib is an intravenous (IV) cyclin-dependent kinase 4/6 (CDK4/6) inhibitor. Preliminary data showed that adding trilaciclib prior to gemcitabine plus carboplatin (GCb) significantly increased overall survival (OS) compared with GCb alone among patients with metastatic triple-negative breast cancer (mTNBC) (Tan et al., Lancet Oncol. 2019;20:1587-1601). Here, final antitumor efficacy results (objective response rate [ORR], progression-free survival [PFS], and OS) are reported for the whole study population, and in cohorts according to CDK4/6 dependence and level of programmed death ligand-1 (PD-L1) expression. Methods This was a randomized, open-label, phase 2 study of patients with mTNBC who had received ≤2 previous lines of chemotherapy in the recurrent/metastatic setting (NCT02978716). Patients were randomized (1:1:1) to receive GCb on days 1 and 8 (group 1, n=34), trilaciclib prior to GCb on days 1 and 8 (group 2, n=33), or trilaciclib alone on days 1 and 8 and prior to GCb on days 2 and 9 (group 3, n=35), in 21-day cycles. PFS and OS (prespecified secondary endpoints) were assessed in the intention-to-treat (ITT) population, and ORR in response-evaluable patients. Patient tumors were characterized as CDK4/6 independent (basal-like) or indeterminate (HER2-enriched, normal-like, luminal A/B) according to the established PAM50 signature, or CDK4/6 dependent (luminal androgen receptor) or indeterminate (basal-like 1/2, mesenchymal) according to the established Lehmann signature. PD-L1 expression was scored as negative or positive if Results Median follow-up was 8.4 months (range: 0.1-25.7) for group 1, 14.0 months (1.3-33.6) for group 2, and 15.3 months (3.5-33.7) for group 3. The ORR among response-evaluable patients was 7/24 (29.2%) in group 1, 15/30 (50.0%) in group 2, and 12/31 (38.7%) in group 3. Median PFS (95% confidence interval [CI]) in the ITT population was 5.7 (3.3, 9.9) months in group 1, 9.4 (6.1, 11.9) months in group 2, and 7.3 (6.2, 13.9) months in group 3, with hazard ratios (HRs) of 0.62 (P = 0.2099) and 0.63 (P = 0.1816), for groups 2 and 3 versus group 1, respectively. Overall, 73.5%, 39.4%, and 57.1% of patients in groups 1, 2, and 3 had died. Median OS (95% CI) was 12.6 (6.3, 15.6) months in group 1, not reached (NR) (10.2, NR) in group 2 (HR = 0.31, P = 0.0016), and 17.8 (12.9, 32.7) months in group 3 (HR = 0.40, P = 0.0004). For groups 2 and 3 combined, median OS was 19.8 (14.0, NR) months (HR = 0.37, P Conclusions Mature data from this study confirm that administering trilaciclib prior to GCb enhances antitumor efficacy compared with GCb alone, with statistically significant improvements in OS. Subgroup analyses suggest that adding trilaciclib prior to GCb benefits patients regardless of CDK4/6 dependence status and PD-L1 expression. Additional immune subtyping analyses are ongoing and will be presented. Group 1Group 2Group 3PD-L1 +vePD-L1 –vePD-L1 +vePD-L1 –vePD-L1 +vePD-L1 –vePatients, n171016101616ORR, n (%)4 (23.5)3 (30.0)8 (50.0)4 (40.0)7 (43.8)4 (25.0)Median PFS, months (95% CI)3.5 (2.2, NR)9.5 (5.2, NR)7.9 (4.3, NR)11.9 (8.8, NR)9.0 (6.2, NR)6.9 (6.4, NR)P value (Wald Test)––0.3470.6040.0690.766HR (95% CI)––0.70 (0.3, 1.5)0.76 (0.3, 2.2)0.46 (0.2, 1.1)1.16 (0.4, 3.1)Median OS, months (95% CI)10.5 (6.3, 18.8)13.9 (12.6, NR)20.1 (10.2, NR)NR (9.4, NR)32.7 (15.3, NR)17.8 (12.9, NR)P value (Wald Test)––0.0280.0830.020.239HR (95% CI)––0.35 (0.1, 0.9)0.34 (0.1, 1.2)0.33 (0.1, 0.8)0.57 (0.2, 1.5)HR and P values are for comparisons between group 2 versus group 1, and group 3 versus group 1.+ve, positive; –ve, negative; CI, confidence interval; HR, hazard ratio; NR, not reached; ORR, objective response rate; OS, overall survival; PD-L1, programmed death ligand-1; PFS, progression-free survival. Citation Format: Joyce O'Shaughnessy, Gail S Wright, Anu R Thummala, Michael A Danso, Lazar Popovic, Timothy J Pluard, Hyo S Han, Željko Vojnović, Nikola Vasev, Ling Ma, Donald A Richards, Sharon T Wilks, Dušan Milenković, Jie Xiao, Jessica A Sorrentino, Janet Horton, Antoinette R Tan. Trilaciclib improves overall survival when given with gemcitabine/carboplatin in patients with metastatic triple-negative breast cancer: Final analysis of a randomized phase 2 trial [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PD1-06.
- Published
- 2021
21. Pembrolizumab versus placebo as post-nephrectomy adjuvant therapy for clear cell renal cell carcinoma (KEYNOTE-564): 30-month follow-up analysis of a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial
- Author
-
Thomas Powles, Piotr Tomczak, Se Hoon Park, Balaji Venugopal, Thomas Ferguson, Stefan N Symeonides, Jaroslav Hajek, Howard Gurney, Yen-Hwa Chang, Jae Lyun Lee, Naveed Sarwar, Antoine Thiery-Vuillemin, Marine Gross-Goupil, Mauricio Mahave, Naomi B Haas, Piotr Sawrycki, Joseph E Burgents, Lei Xu, Kentaro Imai, David I Quinn, Toni K Choueiri, Toni Choueiri, Thomas R. Ferguson, Tzu-Ping Lin, Stefan N. Symeonides, Naomi B. Haas, Howard P. Gurney, Christine Chevreau, John M. Burke, Gurjyot Doshi, Bohuslav Melichar, Delphine Topart, Stephane Oudard, Evgeniy Kopyltsov, Hans-Joerg Hammers, David I. Quinn, Ajjai Alva, Juliana de Janoski Menezes, Adriano Goncalves e Silva, Eric W. Winquist, Alketa Hamzaj, Giuseppe Procopio, Boguslawa Karaszewska, Ewa M. Nowakowska-Zajdel, Boris Y. Alekseev, Rustem A. Gafanov, Adel Izmailov, Andrey Semenov, Sergey G. Afanasyev, Oleg N. Lipatov, Thomas B. Powles, Sandy Srinivas, David McDermott, Samith T. Kochuparambil, Ian D. Davis, Katriina Peltola, Roberto Sabbatini, Jinsoo Chung, Michail I. Shkolnik, Vsevolod B. Matveev, Pablo Gajate Borau, Steven McCune, Thomas E. Hutson, Alejandro Dri, Silvio Correia Sales, Carrie Yeung, Carmen Marcela Alcala Castro, Peter Bostrom, Brigitte Laguerre, Consuelo Buttigliero, Ugo de Giorgi, Eugeniy A. Fomin, Yousef Zakharia, Clara Hwang, Eric A. Singer, Jeffrey T. Yorio, David Waterhouse, Ruben Dario Kowalyszyn, Margarita Sonia Alfie, Eduardo Yanez Ruiz, Tomas Buchler, Krista Kankaanranta, Gianluigi Ferretti, Go Kimura, Kazuo Nishimura, Naoya Masumori, Satoshi Tamada, Haruaki Kato, Hiroshi Kitamura, Iwona Danielewicz, Joanna Wojcik-Tomaszewska, Nuria Sala Gonzalez, Kun-Yuan Chiu, Michael B. Atkins, Elisabeth Heath, Gustavo Adolfo Rojas-Uribe, Manuel Enrique Gonzalez Fernandez, Susan Feyerabend, Sandro Pignata, Kazuyuki Numakura, Bozena Cybulska Stopa, Ruslan Zukov, Miguel Angel Climent Duran, Pablo Jose Maroto Rey, Alvaro Montesa Pino, Chao-Hsiang Chang, Salil Vengalil, Tom S. Waddell, Patrick W. Cobb, Ralph Hauke, Daniel M. Anderson, John Sarantopoulos, Theodore Gourdin, Tian Zhang, Gautam Jayram, Luis Enrique Fein, Carole Harris, Patricia Medeiros Milhomem Beato, Francisco Flores, Angela Estay, Juan Andres Rubiano, Jens Bedke, Stefan Hauser, Andreas Neisius, Jonas Busch, Satoshi Anai, Hiroyuki Tsunemori, Dariusz Sawka, Bozena Sikora-Kupis, Jose Angel Arranz, Ignacio Delgado, Chung-Hsin Chen, Elizabeth Gunderson, Scott Tykodi, Alan Koletsky, Kevin Chen, Manish Agrawal, Diego Lucas Kaen, Juan Pablo Sade, Marcelo Daniel Tatangelo, Francis Parnis, Fernando Maciel Barbosa, Genevieve Faucher, Nayyer Iqbal, Daniele Marceau, Jean-Benoit Paradis, Nawar Hanna, Alejandro Acevedo, Carolina Ibanez, Luis Villanueva, Pedro Pablo Galaz, Isabel Cristina Durango, Ray Manneh, Zdenek - Kral, Petra Holeckova, Heikki Hakkarainen, Hanna Ronkainen, Sophie Abadie-Lacourtoisie, Sophie Tartas, Peter J. Goebell, Marc-Oliver Grimm, Thomas Hoefner, Manfred Wirth, Andrej Panic, Wolfgang Schultze-Seemann, Akira Yokomizo, Ryuichi Mizuno, Hirotsugu Uemura, Masatoshi Eto, Masao Tsujihata, Yoshihisa Matsukawa, Yoji Murakami, Miso Kim, Paul Hamberg, Malgorzata Marczewska-Skrodzka, Cezary Szczylik, Alison C. Humphreys, Peter Jiang, Birendra Kumar, Gary Lu, Arpita Desai, Jose Antonio Karam, George Keogh, Mark Fleming, Juan Jose Zarba, Viviana E. Leiva, Guillermo Ariel Mendez, Samuel J. Harris, Stephen J. Brown, Joao Neif Antonio Junior, Rita de Cassia Costamilan, Roberto Odebrecht Rocha, David Muniz, Leandro Brust, Aly-Khan Lalani, Jeffrey Graham, Michael Levesque, Francisco Orlandi, Rostislav Kotasek, Jean L. Deville, Delphine Borchiellini, Axel Merseburger, Michael Rink, Frederik Roos, Ray McDermott, Masafumi Oyama, Yoshiaki Yamamoto, Yoshihiko Tomita, Yuji Miura, Naomasa Ioritani, Hans Westgeest, Tomasz Kubiatowski, Wieslaw Bal, Regina Girones Sarrio, Julie Rowe, Debra M. Prow, Francis Senecal, Neda Hashemi-Sadraei, Scott W. Cole, Stephan D. Kendall, Donald A. Richards, Ian D. Schnadig, and Mukul Gupta
- Subjects
Adult ,Oncology ,Double-Blind Method ,Antineoplastic Combined Chemotherapy Protocols ,Hypertension ,Humans ,Antibodies, Monoclonal, Humanized ,Carcinoma, Renal Cell ,Nephrectomy ,Kidney Neoplasms ,Follow-Up Studies - Abstract
Background: The first interim analysis of the KEYNOTE-564 study showed improved disease-free survival with adjuvant pembrolizumab compared with placebo after surgery in patients with clear cell renal cell carcinoma at an increased risk of recurrence. The analysis reported here, with an additional 6 months of follow-up, was designed to assess longer-term efficacy and safety of pembrolizumab versus placebo, as well as additional secondary and exploratory endpoints. Methods: In the multicentre, randomised, double-blind, placebo-controlled, phase 3 KEYNOTE-564 trial, adults aged 18 years or older with clear cell renal cell carcinoma with an increased risk of recurrence were enrolled at 213 hospitals and cancer centres in North America, South America, Europe, Asia, and Australia. Eligible participants had an Eastern Cooperative Oncology Group performance status of 0 or 1, had undergone nephrectomy 12 weeks or less before randomisation, and had not received previous systemic therapy for advanced renal cell carcinoma. Participants were randomly assigned (1:1) via central permuted block randomisation (block size of four) to receive pembrolizumab 200 mg or placebo intravenously every 3 weeks for up to 17 cycles. Randomisation was stratified by metastatic disease status (M0 vs M1), and the M0 group was further stratified by ECOG performance status and geographical region. All participants and investigators involved in study treatment administration were masked to the treatment group assignment. The primary endpoint was disease-free survival by investigator assessment in the intention-to-treat population (all participants randomly assigned to a treatment). Safety was assessed in the safety population, comprising all participants who received at least one dose of pembrolizumab or placebo. As the primary endpoint was met at the first interim analysis, updated data are reported without p values. This study is ongoing, but no longer recruiting, and is registered with ClinicalTrials.gov, NCT03142334. Findings: Between June 30, 2017, and Sept 20, 2019, 994 participants were assigned to receive pembrolizumab (n=496) or placebo (n=498). Median follow-up, defined as the time from randomisation to data cutoff (June 14, 2021), was 30·1 months (IQR 25·7–36·7). Disease-free survival was better with pembrolizumab compared with placebo (HR 0·63 [95% CI 0·50–0·80]). Median disease-free survival was not reached in either group. The most common all-cause grade 3–4 adverse events were hypertension (in 14 [3%] of 496 participants) and increased alanine aminotransferase (in 11 [2%]) in the pembrolizumab group, and hypertension (in 13 [3%] of 498 participants) in the placebo group. Serious adverse events attributed to study treatment occurred in 59 (12%) participants in the pembrolizumab group and one (
- Published
- 2022
22. A Phase Ib multicenter, dose-escalation study of the polyamine analogue PG-11047 in combination with gemcitabine, docetaxel, bevacizumab, erlotinib, cisplatin, 5-fluorouracil, or sunitinib in patients with advanced solid tumors or lymphoma
- Author
-
Carlos Becerra, Donald A. Richards, Thomas E. Boyd, Paul Conkling, Hillary H. Wu, Tracy Murray Stewart, Joe Stephenson, Daniel D. Von Hoff, Nicholas J. Vogelzang, Robert A. Casero, Lawrence Garbo, David Smith, Michael L. Fitzgerald, Robert M. Jotte, and Laurence J. Marton
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lymphoma ,Maximum Tolerated Dose ,Bevacizumab ,medicine.medical_treatment ,Docetaxel ,Toxicology ,Deoxycytidine ,Article ,Erlotinib Hydrochloride ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Sunitinib ,medicine ,Humans ,Pharmacology (medical) ,Aged ,Aged, 80 and over ,Pharmacology ,Cisplatin ,Chemotherapy ,Dose-Response Relationship, Drug ,business.industry ,Middle Aged ,Gemcitabine ,030104 developmental biology ,Fluorouracil ,030220 oncology & carcinogenesis ,Female ,Spermine ,Erlotinib ,business ,medicine.drug - Abstract
PURPOSE: Polyamines are absolutely essential for maintaining tumor cell proliferation. PG-11047, a polyamine analogue, is a nonfunctional competitor of the natural polyamine spermine that has demonstrated anticancer activity in cells and animal models of multiple cancer types. Preclinical investigations into the effects of common chemotherapeutic agents have revealed overlap with components of the polyamine metabolic pathway also affected by PG-11047. This report describes a Phase Ib clinical trial investigating PG-11047 in combination with cytotoxic and anti-angiogenic chemotherapeutic agents in patients with advanced refractory metastatic solid tumors or lymphoma. METHODS: A total of 172 patients were assigned to treatment arms based on cancer type to receive the appropriate standard-of-care therapy (gemcitabine, docetaxel, bevacizumab, erlotinib, cisplatin, 5-fluorouracil (5-FU), or sunitinib as directed) along with intravenous infusions of PG-11047 on days 1, 8, and 15 of a 28-day cycle. PG-11047 dose escalation ranged from 50 mg to 590 mg. RESULTS: The maximum tolerated dose (MTD) of PG-11047 in combination with bevacizumab, erlotinib, cisplatin, and 5-FU was 590 mg. Dose-limiting toxicities (DLTs) in these groups were rare (5 of 148 patients). Overall partial responses (PR) were observed in 12% of patients treated with PG-11047 and bevacizumab, with stable disease documented in an additional 40%. Stable disease occurred in 71.4% of patients in the 5-FU arm, 54.1% in the cisplatin arm, and 33.3% in the erlotinib arm. Four of the patients receiving cisplatin + PG-11047 (20%) had unconfirmed PRs. MTDs for gemcitabine, docetaxel, and sunitinib could not be determined due to DLTs at low doses of PG-11047 and small sample size. CONCLUSIONS: Results of this Phase Ib trial indicate that PG-11047 can be safely administered to patients in combination with bevacizumab, erlotinib, cisplatin, and 5-FU on the once weekly dosing schedule described and may provide therapeutic benefit. The manageable toxicity profile and high MTD determination provide a safety profile for further clinical studies.
- Published
- 2020
23. Randomised phase II trial of gemcitabine and nab-paclitaxel with necuparanib or placebo in untreated metastatic pancreas ductal adenocarcinoma
- Author
-
David P. Ryan, Eileen M. O'Reilly, Kenneth H. Yu, James M. Roach, Donald A. Richards, Spencer H. Shao, Julie Wolf, Tanios Bekaii-Saab, Diletta Barone, Keith T. Flaherty, Devalingam Mahalingam, Molly Rosano, and Silva Krause
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Paclitaxel ,medicine.drug_class ,Low molecular weight heparin ,Placebo ,Deoxycytidine ,Gastroenterology ,Article ,03 medical and health sciences ,Subcutaneous injection ,0302 clinical medicine ,Double-Blind Method ,Albumins ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Data monitoring committee ,Aged ,Aged, 80 and over ,business.industry ,Hazard ratio ,Middle Aged ,Prognosis ,Interim analysis ,Gemcitabine ,Confidence interval ,Pancreatic Neoplasms ,Survival Rate ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,Heparitin Sulfate ,business ,Carcinoma, Pancreatic Ductal ,Follow-Up Studies ,medicine.drug - Abstract
Background Necuparanib, a rationally engineered low-molecular-weight heparin, combined with gemcitabine/nab-paclitaxel showed an encouraging safety and oncologic signal in a phase Ib trial. This randomised multicentre phase II trial evaluates the addition of necuparanib or placebo to gemcitabine/nab-paclitaxel in untreated metastatic pancreatic ductal adenocarcinoma (PDAC). Patients and methods Eligibility included 18 years, histologically or cytologically confirmed metastatic PDAC, measurable disease and Eastern Co-Operative Oncology Group performance status of 0–1. Patients were randomly assigned to necuparanib (5 mg/kg subcutaneous injection once daily) or placebo (subcutaneous injection once daily) and gemcitabine/nab-paclitaxel on days 1, 8 and 15 of 28-day cycles. The primary end-point was median overall survival (OS), and secondary end-points included median progression-free survival, response rates and safety. Results One-hundred ten patients were randomised, 62 to necuparanib arm and 58 to placebo arm. The futility boundary was crossed at a planned interim analysis, and the study was terminated by the Data Safety Monitoring Board. The median OS was 10.71 months (95% confidence interval [CI]: 7.95–11.96) for necuparanib arm and 9.99 months (95% CI: 7.85–12.85) for placebo arm (hazard ratio: 1.12, 95% CI: 0.66–1.89, P-value: 0.671). The necuparanib arm had a higher incidence of haematologic toxicity relative to placebo patients (83% and 70%). Conclusion The addition of necuparanib to standard of care treatment for advanced PDAC did not improve OS. Safety was acceptable. No further development of necuparanib is planned although targeting the coagulation cascade pathway remains relevant in PDAC. NCT01621243
- Published
- 2020
24. Trilaciclib Prior to Chemotherapy in Patients with Metastatic Triple-Negative Breast Cancer: Final Efficacy and Subgroup Analysis from a Randomized Phase II Study
- Author
-
Antoinette R. Tan, Gail S. Wright, Anu R. Thummala, Michael A. Danso, Lazar Popovic, Timothy J. Pluard, Hyo S. Han, Željko Vojnović, Nikola Vasev, Ling Ma, Donald A. Richards, Sharon T. Wilks, Dušan Milenković, Jie Xiao, Jessica Sorrentino, Janet Horton, and Joyce O'Shaughnessy
- Subjects
Antineoplastic Combined Chemotherapy Protocols Humans Pyrimidines / therapeutic use Pyrroles / therapeutic use Triple Negative Breast Neoplasms* / pathology ,Cancer Research ,Pyrimidines ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Pyrroles ,Triple Negative Breast Neoplasms - Abstract
Purpose: We report final antitumor efficacy results from a phase II study of trilaciclib, an intravenous cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, administered prior to gemcitabine plus carboplatin (GCb) in patients with metastatic triple-negative breast cancer (NCT02978716). Patients and Methods: Patients were randomized (1:1:1) to group 1 [GCb (days 1, 8); n = 34], group 2 [trilaciclib prior to GCb (days 1, 8); n = 33], or group 3 [trilaciclib (days 1, 8) and trilaciclib prior to GCb (days 2, 9); n = 35]. Subgroup analyses were performed according to CDK4/6 dependence, level of programmed death-ligand 1 (PD-L1) expression, and RNA-based immune signatures using proportional hazards regression. T-cell receptor (TCR) β CDR3 regions were amplified and sequenced to identify, quantify, and compare the abundance of each unique TCRβ CDR3 at baseline and on treatment. Results: Median overall survival (OS) was 12.6 months in group 1, not reached in group 2 (HR = 0.31; P = 0.0016), 17.8 months in group 3 (HR = 0.40; P = 0.0004), and 19.8 months in groups 2 and 3 combined (HR = 0.37; P < 0.0001). Efficacy outcomes were comparable regardless of cancer CDK4/6 dependence status and immune signatures. Administering trilaciclib prior to GCb prolonged OS irrespective of PD-L1 status but had greater benefit in the PD-L1–positive population. T-cell activation was enhanced in patients receiving trilaciclib. Conclusions: Administering trilaciclib prior to GCb enhanced antitumor efficacy, with significant improvements in OS. Efficacy outcomes in immunologic subgroups and enhancements in T-cell activation suggest these improvements may be mediated via immunologic mechanisms.
- Published
- 2022
25. Evaluation of cell-free DNA approaches for multi-cancer early detection
- Author
-
Arash Jamshidi, Minetta C. Liu, Eric A. Klein, Oliver Venn, Earl Hubbell, John F. Beausang, Samuel Gross, Collin Melton, Alexander P. Fields, Qinwen Liu, Nan Zhang, Eric T. Fung, Kathryn N. Kurtzman, Hamed Amini, Craig Betts, Daniel Civello, Peter Freese, Robert Calef, Konstantin Davydov, Saniya Fayzullina, Chenlu Hou, Roger Jiang, Byoungsok Jung, Susan Tang, Vasiliki Demas, Joshua Newman, Onur Sakarya, Eric Scott, Archana Shenoy, Seyedmehdi Shojaee, Kristan K. Steffen, Virgil Nicula, Tom C. Chien, Siddhartha Bagaria, Nathan Hunkapiller, Mohini Desai, Zhao Dong, Donald A. Richards, Timothy J. Yeatman, Allen L. Cohn, David D. Thiel, Donald A. Berry, Mohan K. Tummala, Kristi McIntyre, Mikkael A. Sekeres, Alan Bryce, Alexander M. Aravanis, Michael V. Seiden, and Charles Swanton
- Subjects
Chemical Biology & High Throughput ,Signalling & Oncogenes ,Human Biology & Physiology ,Cancer Research ,Ecology,Evolution & Ethology ,Oncology ,Genome Integrity & Repair ,Tumour Biology ,Genetics & Genomics ,Computational & Systems Biology - Abstract
In the Circulating Cell-free Genome Atlas (NCT02889978) substudy 1, we evaluate several approaches for a circulating cell-free DNA (cfDNA)-based multi-cancer early detection (MCED) test by defining clinical limit of detection (LOD) based on circulating tumor allele fraction (cTAF), enabling performance comparisons. Among 10 machine-learning classifiers trained on the same samples and independently validated, when evaluated at 98% specificity, those using whole-genome (WG) methylation, single nucleotide variants with paired white blood cell background removal, and combined scores from classifiers evaluated in this study show the highest cancer signal detection sensitivities. Compared with clinical stage and tumor type, cTAF is a more significant predictor of classifier performance and may more closely reflect tumor biology. Clinical LODs mirror relative sensitivities for all approaches. The WG methylation feature best predicts cancer signal origin. WG methylation is the most promising technology for MCED and informs development of a targeted methylation MCED test.
- Published
- 2022
26. Myeloprotective Effects of Trilaciclib Among Patients with Small Cell Lung Cancer at Increased Risk of Chemotherapy-Induced Myelosuppression: Pooled Results from Three Phase 2, Randomized, Double-Blind, Placebo-Controlled Studies
- Author
-
Donald A. Richards, J. Thaddeus Beck, Rajesh K. Malik, Joyce M Antal, Ivan Sinielnikov, Keith Lerro, Marielle Sabatini, Maen A. Hussein, Marina Maglakelidze, Alexander I. Spira, Todd A Gersten, and Yili Pritchett
- Subjects
myelosuppression ,Chemotherapy ,medicine.medical_specialty ,trilaciclib ,business.industry ,Anemia ,medicine.medical_treatment ,Incidence (epidemiology) ,Subgroup analysis ,Placebo ,medicine.disease ,chemotherapy ,Clinical trial ,Oncology ,Cancer Management and Research ,Internal medicine ,medicine ,Patient-reported outcome ,small cell lung cancer ,myelopreservation ,business ,myeloprotection ,Febrile neutropenia ,Original Research - Abstract
Maen Hussein,1 Marina Maglakelidze,2 Donald A Richards,3 Marielle Sabatini,4 Todd A Gersten,5 Keith Lerro,6 Ivan Sinielnikov,7 Alexander Spira,8,9 Yili Pritchett,10 Joyce M Antal,10 Rajesh Malik,10 J Thaddeus Beck11 1Florida Cancer Specialists, Leesburg, FL, USA; 2LLC Arensia Exploratory Medicine, Tbilisi, Georgia; 3Texas Oncology-Tyler, US Oncology Research, Tyler, TX, USA; 4Saint Leon Hospital, Bayonne, France; 5Florida Cancer Specialists, West Palm Beach, FL, USA; 6Regional Medical Oncology Center, Wilson, NC, USA; 7Volyn Regional Oncology Center, Lutsk, Ukraine; 8Virginia Cancer Specialists, Fairfax, VA, USA; 9US Oncology Research, The Woodlands, TX, USA; 10G1 Therapeutics, Inc., Research Triangle Park, NC, USA; 11Highlands Oncology Group, Fayetteville, AR, USACorrespondence: Maen HusseinFlorida Cancer Specialists, Leesburg, FL, USATel +1 352-787-9448Email mhussein@flcancer.comPurpose: Trilaciclib is an intravenous cyclin-dependent kinase 4/6 inhibitor indicated to decrease the incidence of chemotherapy-induced myelosuppression (CIM) by protecting hematopoietic stem and progenitor cells and immune system function from chemotherapy-induced damage (myeloprotection). Here, we investigated the myeloprotective effects of trilaciclib among patients at increased risk of CIM.Patients and Methods: Data were pooled from three randomized, double-blind, placebo-controlled, phase 2 clinical studies of trilaciclib administered prior to chemotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC). Myeloprotective outcomes were evaluated in patient subgroups based on age (< 65 or ≥ 65 years), risk of chemotherapy-induced febrile neutropenia (FN), and risk of anemia or red blood cell (RBC) transfusions. For the FN and anemia analyses, risk factors were identified from published literature and used to classify patients into FN and anemia risk categories. Subgroup analysis based on age was also performed on patient reported outcome (PRO) measures.Results: In total, 123 patients received trilaciclib and 119 patients received placebo. Myeloprotective benefits of trilaciclib were observed regardless of age, with greater effects observed among patients aged ≥ 65 years. Across FN risk factors and categories, trilaciclib had beneficial effects on neutrophil-related endpoints vs placebo, with greater effects observed in patients at higher risk of FN. Effects on RBC-related endpoints favored trilaciclib vs placebo, regardless of anemia risk factors and categories. Improvements in PROs with trilaciclib were observed irrespective of age group, but with greater improvements and less deterioration from baseline observed in older patients.Conclusion: By both decreasing the incidence of CIM and improving quality of life, trilaciclib has the potential to allow patients receiving chemotherapy for ES-SCLC, including patients who are older or more vulnerable to CIM, to receive chemotherapy on schedule and at standard-of-care doses, and to improve the experience for patients receiving chemotherapy to treat ES-SCLC.Clinical Trial Numbers: NCT02499770; NCT03041311; NCT02514447.Keywords: trilaciclib, myelosuppression, myeloprotection, myelopreservation, chemotherapy, small cell lung cancer
- Published
- 2021
27. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set
- Author
-
Rosanna Lapham, Charles Swanton, Jianjun Gao, David Cosgrove, Eric A. Klein, Allen Lee Cohn, G. Chung, Nathan Hunkapiller, Donald A. Richards, Kathryn N. Kurtzman, Jessica M. Clement, Minetta C. Liu, Arash Jamshidi, Michael V. Seiden, and M.K. Tummala
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Stage ii ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Neoplasms ,Biomarkers, Tumor ,Medicine ,Humans ,Prospective Studies ,Liquid biopsy ,Stage (cooking) ,Early Detection of Cancer ,business.industry ,Cancer ,Hematology ,Oncogenes ,DNA Methylation ,medicine.disease ,Cancer Early Detection ,Confidence interval ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,business ,Stage iv - Abstract
Background A multi-cancer early detection (MCED) test used to complement existing screening could increase the number of cancers detected through population screening, potentially improving clinical outcomes. The Circulating Cell-free Genome Atlas study (CCGA; NCT02889978) was a prospective, case-controlled, observational study and demonstrated that a blood-based MCED test utilizing cell-free DNA (cfDNA) sequencing in combination with machine learning could detect cancer signals across multiple cancer types and predict cancer signal origin (CSO) with high accuracy. The objective of this third and final CCGA substudy was to validate an MCED test version further refined for use as a screening tool. Patients and methods This pre-specified substudy included 4077 participants in an independent validation set (cancer: n = 2823; non-cancer: n = 1254, non-cancer status confirmed at year-one follow-up). Specificity, sensitivity, and CSO prediction accuracy were measured. Results Specificity for cancer signal detection was 99.5% [95% confidence interval (CI): 99.0% to 99.8%]. Overall sensitivity for cancer signal detection was 51.5% (49.6% to 53.3%); sensitivity increased with stage [stage I: 16.8% (14.5% to 19.5%), stage II: 40.4% (36.8% to 44.1%), stage III: 77.0% (73.4% to 80.3%), stage IV: 90.1% (87.5% to 92.2%)]. Stage I-III sensitivity was 67.6% (64.4% to 70.6%) in 12 pre-specified cancers that account for approximately two-thirds of annual USA cancer deaths and was 40.7% (38.7% to 42.9%) in all cancers. Cancer signals were detected across >50 cancer types. Overall accuracy of CSO prediction in true positives was 88.7% (87.0% to 90.2%). Conclusion In this pre-specified, large-scale, clinical validation substudy, the MCED test demonstrated high specificity and accuracy of CSO prediction and detected cancer signals across a wide diversity of cancers. These results support the feasibility of this blood-based MCED test as a complement to existing single-cancer screening tests. Clinical trial number NCT02889978.
- Published
- 2021
28. Performance of a Multi-Cancer Detection Test as a Tool for Diagnostic Resolution of Symptomatic Gynecological Cancers
- Author
-
Minetta C. Liu, David D. Thiel, Jianjun Gao, Eric A. Klein, C. Becerra, Donald A. Richards, E.T. Fung, Earl Hubbell, Kathryn N. Kurtzman, T. Wu, Q. Zhang, X. Chen, Arash Jamshidi, Alan H. Bryce, N. Zhang, and Michael V. Seiden
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Endometriosis ,Obstetrics and Gynecology ,Cancer ,Test sensitivity ,Subgroup analysis ,Disease ,Cancer detection ,medicine.disease ,Confidence interval ,Internal medicine ,medicine ,Stage (cooking) ,business - Abstract
Study Objective Assess performance of a multi-cancer detection (MCD) test. Design Circulating Cell-free Genome Atlas (CCGA; NCT02889978) is a prospective, longitudinal, case-control study. Samples from the second CCGA substudy were used to validate performance of an MCD test, which uses targeted methylation-based cell-free DNA sequencing and machine learning classifiers to detect cancer signal, including from clinically presenting gynecologic cancers, and predict cancer signal origin (CSO). Setting N/A. Patients or Participants MCD test sensitivity and CSO prediction accuracy were evaluated in a subgroup of participants with clinically presenting cancers (CPCs). Specificity was assessed in noncancer participants and a subgroup with significant nonmalignant conditions, including endometriosis. Interventions N/A. Measurements and Main Results Specificity was 99.5% (95% confidence interval [CI]: 98.2-99.9%; 396/398) for the noncancer group and 93.8% (71.7-99.7%; 15/16) for the significant nonmalignant conditions noncancer subgroup. Overall sensitivity was 66.4% (62.2-70.3%; 344/518) for CPC participants, 50.0% (15.0-85.0%; 2/4) for cervical, 70.6% (46.9-86.7%; 12/17) for ovarian, and 25.0% (13.8-41.1%; 9/36) for uterine cancers and varied by stage (Table). CSO prediction accuracy was broadly consistent with CSOs for CPC participants with cancers detected (excluding those with multiple, unknown primaries, or “other” cancer; 91.7% [88.3-94.3%; 300/327]). Conclusion Consistent with the overall CCGA study, this MCD test performed with high specificity and accuracy of CSO prediction in this subgroup analysis of CPCs, including gynecological cancers. Findings support potential use of this test for diagnostic workup of symptomatic disease.
- Published
- 2021
29. A Phase 2 Randomized Placebo-Controlled Adjuvant Trial of GI-4000, a Recombinant Yeast Expressing Mutated RAS Proteins in Patients with Resected Pancreas Cancer
- Author
-
Timothy C. Rodell, Claire Coeshott, Kristi McIntyre, Joanna Roder, Heinrich Roder, David Apelian, Frank E. Harrell, Allen Lee Cohn, Tanios Bekaii-Saab, William E. Fisher, Alexander S. Rosemurgy, Donald A. Richards, Peter Muscarella, Alicia Mattson, Sharona Ross, and Patrick J. Flynn
- Subjects
business.industry ,medicine.medical_treatment ,Cancer ,clinical trial ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Placebo ,medicine.disease ,T cell response ,lcsh:RC254-282 ,Recombinant yeast ,Clinical trial ,medicine.anatomical_structure ,pancreas cancer ,medicine ,Cancer research ,In patient ,Original Article ,immunotherapy ,Pancreas ,business ,K-ras ,Adjuvant - Abstract
Purpose: GI-4000, a series of recombinant yeast expressing four different mutated RAS proteins, was evaluated in subjects with resected ras-mutated pancreas cancer. Methods: Subjects (n?=?176) received GI-4000 or placebo plus gemcitabine. Subjects' tumors were genotyped to identify which matched GI-4000 product to administer. Immune responses were measured by interferon-? (IFN?) ELISpot assay and by regulatory T cell (Treg) frequencies on treatment. Pretreatment plasma was retrospectively analyzed by matrix-assisted laser desorption/ionization-time-of-flight (MALDI-ToF) mass spectrometry for proteomic signatures predictive of GI-4000 responsiveness. Results: GI-4000 was well tolerated, with comparable safety findings between treatment groups. The GI-4000 group showed a similar pattern of median recurrence-free and overall survival (OS) compared with placebo. For the prospectively defined and stratified R1 resection subgroup, there was a trend in 1 year OS (72% vs. 56%), an improvement in OS (523.5 vs. 443.5 days [hazard ratio (HR)?=?1.06 [confidence interval (CI): 0.53?2.13], p?=?0.872), and increased frequency of immune responders (40% vs. 8%; p?=?0.062) for GI-4000 versus placebo and a 159-day improvement in OS for R1 GI-4000 immune responders versus placebo (p?=?0.810). For R0 resection subjects, no increases in IFN? responses in GI-4000?treated subjects were observed. A higher frequency of R0/R1 subjects with a reduction in Tregs (CD4+/CD45RA+/Foxp3low) was observed in GI-4000?treated subjects versus placebo (p?=?0.033). A proteomic signature was identified that predicted response to GI-4000/gemcitabine regardless of resection status. Conclusion: These results justify continued investigation of GI-4000 in studies stratified for likely responders or in combination with immune check-point inhibitors or other immunomodulators, which may provide optimal reactivation of antitumor immunity. ClinicalTrials.gov Number: NCT00300950.
- Published
- 2021
30. 394 Interleukin-8–neutralizing monoclonal antibody BMS-986253 plus nivolumab (NIVO) in biomarker-enriched, primarily anti–PD-(L)1–experienced patients with advanced cancer: initial phase 1 results
- Author
-
Vinit Kumar, Ignacio Melero, Donald A. Richards, Martin Gutierrez, Matthew Dallos, Matteo Simonelli, Janaki Parameswaran, Sarina Anne Piha-Paul, Santanu Dutta, Xiaochen Zhao, Diwakar Davar, Emiliano Calvo, and Jason M. Melear
- Subjects
medicine.medical_specialty ,Nausea ,business.industry ,Melanoma ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Rash ,Gastroenterology ,Pharmacokinetics ,Pharmacodynamics ,Internal medicine ,medicine ,medicine.symptom ,Nivolumab ,Lung cancer ,business - Abstract
Background Interleukin 8 (IL-8) is a C-X-C chemokine that exerts protumorigenic effects in the tumor microenvironment, including recruiting immunosuppressive PMN-MDSCs and promoting angiogenesis.1–3 Elevated serum IL-8 (sIL-8) is a negative prognostic indicator in patients with solid tumors and may have predictive value in patients treated with immunotherapies.2 4 5 BMS-986253, a fully human-sequence IgG1κ anti–IL-8 monoclonal antibody, binds IL-8 and prevents signaling through CXCR1/CXCR2 and has been shown to be safe in patients with advanced cancers.3 We present initial results of BMS-986253 + NIVO from a phase 1/2a trial in patients with advanced cancers who had detectable sIL-8 levels, the majority of whom had progressed on/after prior anti–PD-(L)1 (NCT03400332). Methods During safety evaluation/dose exploration, patients with advanced metastatic solid tumors (melanoma, NSCLC, SCCHN, RCC, or UCC) and detectable sIL-8 (>10 pg/mL at screening) received BMS-986253 600 (n=16), 1200 (n=15), or 2400 mg (n=18) Q4W, or 1200 (n=12) or 2400 mg (n=59) Q2W, + NIVO 480 mg intravenously Q4W. Safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity were evaluated (investigator-assessed, RECIST v1.1). Results As of March 20, 2020, 120 patients (median age, 63 years [range, 35–87 years]) received BMS-986253 + NIVO; 97% of patients received prior anti–PD-(L)1 therapy, and 25% received prior anti–CTLA-4 therapy. BMS-986253 + NIVO was well tolerated with no dose-limiting toxicities observed. Most TRAEs were grade 1–2. The most common (≥5% of patients) TRAEs (any grade; grade 3–4) were fatigue (9%; 1%), nausea (7%; 0%), rash/rash maculopapular (6%; 0%), pruritus (5%; 0%), and decreased appetite (5%; 0%). Grade 3–4 serious TRAEs were reported in 2 patients (infusion-related reaction, BMS-986253 2400 mg Q2W + NIVO; AST/ALT increased, BMS-986253 1200 mg Q4W + NIVO). BMS-986253 exposure increased dose proportionally and was not altered with NIVO. BMS-986253 resulted in dose-dependent reductions in free sIL-8 levels, with tumor IL-8 suppression detected in most patients evaluated; additional pharmacodynamic endpoints will be presented. Partial responses were observed in multiple tumor types, including 5 of 28 patients with melanoma who had progressed on/after prior anti–PD-(L)1; 4 of the 5 patients were also previously treated with anti–CTLA-4. Conclusions BMS-986253 + NIVO demonstrated a tolerable safety profile with dose-proportional pharmacokinetics and robust sIL-8 suppression. Preliminary antitumor activity was observed across a range of doses/regimens in this biomarker-enriched, anti–PD-(L)1–experienced, heterogeneous patient population with advanced cancers. These findings support further evaluation of BMS-986253 in select advanced tumors. Acknowledgements The authors acknowledge Dr Charles Drake while at Columbia University Medical Center, New York, NY, USA, for his contributions to the study. Trial Registration NCT03400332 Ethics Approval This study was approved by the WCG Independent Review Board, approval number 20172711. References David JM, Dominguez C, Hamilton DH, et al. The IL-8/IL-8R axis: a double agent in tumor immune resistance. Vaccines (Basel) 2016;4:22. Schalper KA, Carleton M, Zhou M, et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 2020;26:688–692. Bilusic M, Heery CR, Collin JM, et al. Phase I trial of HuMax-IL-8 (BMS-986253), an anti–IL-8 monoclonal antibody, in patients with metastatic or unresectable solid tumors. J Immunother Cancer 2019;7:240. Yuen KC, Liu L-F, Gupta V, et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med 2020;26:683–698. Sanmamed MF, Perez-Gracia JL, Schalper KA, et al. Changes in serum interleukin-8 (IL-8) levels reflect and predict response to anti–PD-1 treatment in melanoma and non-small-cell lung cancer patients. Ann Oncol 2017;28:1988–1995.
- Published
- 2020
31. A New Sophomore Engineering Curriculum The Rose Hulman Experience
- Author
-
Donald E. Richards
- Published
- 2020
32. The Political Economy of TRIPS
- Author
-
Donald G. Richards
- Subjects
Political economy ,Economics ,TRIPS architecture - Published
- 2020
33. Introduction and Background
- Author
-
Donald G. Richards
- Published
- 2020
34. Capital, Class, and the State in the Global Economy
- Author
-
Donald G. Richards
- Subjects
Class (computer programming) ,State (polity) ,media_common.quotation_subject ,Capital (economics) ,Economics ,Economic system ,media_common - Published
- 2020
35. Intellectual Property and Positivist Economic Science
- Author
-
Donald G. Richards
- Subjects
Economic science ,Sociology ,Intellectual property ,Positive economics ,Positivism - Published
- 2020
36. Intellectual Property Rights and the Agricultural Biotechnology Revolution
- Author
-
Donald G. Richards
- Subjects
Political science ,Intellectual property ,Agricultural biotechnology ,Law and economics - Published
- 2020
37. Counterhegemony and the Future of TRIPS
- Author
-
Donald G. Richards
- Subjects
Geography ,TRIPS architecture ,Marketing - Published
- 2020
38. The Ideology of Intellectual Property
- Author
-
Donald G. Richards
- Subjects
media_common.quotation_subject ,Political science ,Ideology ,Intellectual property ,media_common ,Law and economics - Published
- 2020
39. TRIPS and the Global Pharmaceutical Industry
- Author
-
Donald G. Richards
- Subjects
Commerce ,business.industry ,TRIPS architecture ,Business ,Pharmaceutical industry - Published
- 2020
40. Intellectual Property Rights and Global Capitalism
- Author
-
Donald G. Richards
- Published
- 2020
41. Phase Ib clinical trial of the anti-frizzled antibody vantictumab (OMP-18R5) plus paclitaxel in patients with locally advanced or metastatic HER2-negative breast cancer
- Author
-
Randall Henner, Azeez Farooki, Carlos Becerra, Joyce O'Shaughnessy, Shailaja Uttamsingh, Donald A. Richards, Rainer K. Brachmann, Cynthia Osborne, Ann M. Kapoun, Chun Zhang, Jennifer R. Diamond, Lu Xu, Alain C. Mita, Bob Stagg, and Monica M. Mita
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Vantictumab ,Paclitaxel ,Receptor, ErbB-2 ,medicine.medical_treatment ,Breast Neoplasms ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Adverse effect ,Chemotherapy ,business.industry ,Antibodies, Monoclonal ,medicine.disease ,Metastatic breast cancer ,Clinical trial ,030104 developmental biology ,Treatment Outcome ,chemistry ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,business - Abstract
Vantictumab is a monoclonal antibody that binds to frizzled (FZD) receptors and inhibits canonical WNT signaling. This phase Ib dose escalation study enrolled patients with locally recurrent or metastatic HER2-negative breast cancer who were treated with weekly paclitaxel in combination with escalating doses of vantictumab. Patients were enrolled in dose escalation cohorts treated with weekly paclitaxel 90 mg/m2 on days 1, 8 and 15 in combination with vantictumab 3.5–14 mg/kg days 1 and 15 or 3–8 mg/kg day 1 of every 28-day cycle. Primary endpoints were safety, dose-limiting toxicities (DLTs). Secondary endpoints included pharmacokinetics, efficacy and an exploratory biomarker analysis. Forty-eight female patients with a mean age of 54 were enrolled. The majority (66.6%) received prior chemotherapy for recurrent or metastatic disease; 45.8% were hormone receptor (HR)-positive, HER2-negative and 54.2% triple-negative. The most frequent adverse events related to any study treatment were nausea (54.2%), alopecia (52.1%), fatigue (47.9%), and peripheral neuropathy (43.8%). No DLTs occurred; however, 6 patients experienced fractures outside of the DLT window. The overall response rate was 31.3% and the clinical benefit rate was 68.8%. A 6-gene WNT pathway signature showed significant association with progression-free survival (PFS) and overall survival (OS) for the biomarker high versus biomarker low groups (PFS: p = 0.029 and OS: p = 0.00045, respectively). The combination of vantictumab and weekly paclitaxel was generally well tolerated with promising efficacy; however, the incidence of fractures limits future clinical development of this particular WNT inhibitor in metastatic breast cancer. ClinicalTrials.gov registration: NCT01973309
- Published
- 2020
42. Phase Ib Trial of the PI3K Inhibitor Copanlisib Combined with the Allosteric MEK Inhibitor Refametinib in Patients with Advanced Cancer
- Author
-
Carol Peña, Daniel D. Von Hoff, Camille Granvil, Susanne Reschke, George R. Blumenschein, Isabelle Genvresse, Ferry A.L.M. Eskens, Klaus Mross, Donald A. Richards, Adam Skubala, Ramesh K. Ramanathan, and Medical Oncology
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Nausea ,Population ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,Mucositis ,Pharmacology (medical) ,Original Research Article ,education ,Adverse effect ,Copanlisib ,education.field_of_study ,business.industry ,MEK inhibitor ,Correction ,medicine.disease ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Pharmacodynamics ,medicine.symptom ,business - Abstract
Background Dual inhibition of PI3K and MAPK signaling is conceptually a promising anticancer therapy. Objective This phase 1b trial investigated the safety, maximum tolerated dose (MTD), recommended phase II dose, pharmacokinetics, tumor response, fluorodeoxyglucose positron emission tomography (FDG-PET) pharmacodynamics, and biomarker explorations for the combination of pan-PI3K inhibitor copanlisib and allosteric MEK inhibitor refametinib in patients with advanced solid tumors. Patients and methods This was an adaptive trial with eight dose cohorts combining dose escalation and varying schedules in repeated 28-day cycles. Patients received copanlisib (0.2–0.8 mg/kg intravenously) intermittently (days 1, 8, 15) or weekly (days 1, 8, 15, 22) each cycle, and refametinib (30–50 mg twice daily orally) continuously or 4 days on/3 days off. Patients with KRAS, NRAS, BRAF, or PI3KCA mutations were eligible for the expansion cohort. Results In the dose-escalation (n = 49) and expansion (n = 15) cohorts, the most common treatment-emergent adverse events included diarrhea (59.4%), nausea, acneiform rash, and fatigue (51.6% each). Dose-limiting toxicities included oral mucositis (n = 4), increased alanine aminotransferase/aspartate aminotransferase (n = 3), acneiform rash, hypertension (n = 2 each), and diarrhea (n = 1). MTD was copanlisib 0.4 mg/kg weekly and refametinib 30 mg twice daily. No pharmacokinetic interactions were identified. Decreased tumor FDG uptake and MEK-ERK signaling inhibition were demonstrated during treatment. Best response was stable disease (n = 21); median treatment duration was 6 weeks. Conclusions Despite sound rationale and demonstrable pharmacodynamic tumor activity in relevant tumor populations, a dose and schedule could not be identified for this drug combination that were both tolerable and offered clear efficacy in the population assessed. Clinicaltrials.gov identifier NCT01392521. Electronic supplementary material The online version of this article (10.1007/s11523-020-00714-0) contains supplementary material, which is available to authorized users.
- Published
- 2020
43. Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
Thomas J Stout, José Baselga, Philippe L. Bedard, Andrés Cervantes, Donald A. Richards, Jerry Y. Hsu, Na Cui, Maura N. Dickler, Cristina Saura, Manish R. Patel, Ian E. Krop, Mafalda Oliveira, Alison Cardenas, Michael C. Wei, Lajos Pusztai, and Timothy R. Wilson
- Subjects
Adult ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Class I Phosphatidylinositol 3-Kinases ,Receptor, ErbB-2 ,Phases of clinical research ,Breast Neoplasms ,Disease-Free Survival ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Adverse effect ,Fulvestrant ,Aged ,Aged, 80 and over ,Response rate (survey) ,business.industry ,Imidazoles ,Cancer ,Middle Aged ,medicine.disease ,Oxazepines ,030104 developmental biology ,Receptors, Estrogen ,Hormone receptor ,030220 oncology & carcinogenesis ,Mutation ,Toxicity ,Female ,business ,medicine.drug - Abstract
Purpose: This single-arm, open-label phase II study evaluated the safety and efficacy of taselisib (GDC-0032) plus fulvestrant in postmenopausal women with locally advanced or metastatic HER2-negative, hormone receptor (HR)-positive breast cancer.Patients and Methods: Patients received 6-mg oral taselisib capsules daily plus intramuscular fulvestrant (500 mg) until disease progression or unacceptable toxicity. Tumor tissue (if available) was centrally evaluated for PIK3CA mutations. Adverse events (AE) were recorded using NCI-CTCAE v4.0. Tumor response was investigator-determined using RECIST v1.1.Results: Median treatment duration was 4.6 (range: 0.9–40.5) months. All patients experienced ≥1 AE, 30 (50.0%) had grade ≥3 AEs, and 19 (31.7%) experienced 35 serious AEs. Forty-seven of 60 patients had evaluable tissue for central PIK3CA mutation testing [20 had mutations, 27 had no mutation detected (MND)]. In patients with baseline measurable disease, clinical activity was observed in tumors with PIK3CA mutations [best confirmed response rate: 38.5% (5/13; 95% CI, 13.9–68.4); clinical benefit rate (CBR): 38.5% (5/13; 95% CI, 13.9–68.4)], PIK3CA-MND [best confirmed response rate: 14.3% (3/21; 95% CI, 3.0–36.3); CBR: 23.8% (5/21; 95% CI, 8.2–47.2)], and unknown PIK3CA mutation status [best confirmed response rate: 20.0% (2/10; 95% CI, 2.5–55.6); CBR: 30.0% (3/10; 95% CI, 6.7–65.2)].Conclusions: Taselisib plus fulvestrant had clinical activity irrespective of PIK3CA mutation status, with numerically higher objective response rate and CBR in patients with PIK3CA-mutated (vs. -MND) locally advanced or metastatic HER2-negative, HR-positive breast cancer. No new safety signals were reported. A confirmatory phase III trial is ongoing. Clin Cancer Res; 24(18); 4380–7. ©2018 AACR.
- Published
- 2018
44. LY2495655, an antimyostatin antibody, in pancreatic cancer: a randomized, phase 2 trial
- Author
-
Donald A. Richards, Srinivasan Madhusudan, Haofei Tiffany Wang, Boris Lin, Talia Golan, Salomon M. Stemmer, Ravit Geva, and Richard A. Walgren
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Randomization ,Performance status ,business.industry ,Hazard ratio ,medicine.disease ,Placebo ,Cachexia ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Weight loss ,030220 oncology & carcinogenesis ,Physiology (medical) ,Internal medicine ,Pancreatic cancer ,Clinical endpoint ,Medicine ,Orthopedics and Sports Medicine ,medicine.symptom ,business - Abstract
BACKGROUND Cachexia is a formidable clinical challenge in pancreatic cancer. We assessed LY2495655 (antimyostatin antibody) plus standard-of-care chemotherapy in pancreatic cancer using cachexia status as a stratifier. METHODS In this randomized, phase 2 trial, patients with stage II-IV pancreatic cancer were randomized to 300 mg LY2495655, 100 mg LY2495655, or placebo, plus physician-choice chemotherapy from a prespecified list of standard-of-care regimens for first and later lines of care. Investigational treatment was continued during second-line treatment. The primary endpoint was overall survival. RESULTS Overall, 125 patients were randomized. In August 2014, 300 mg LY2495655 was terminated due to imbalance in death rates between the treatment arms; in January 2015, 100 mg LY2495655 treatment was terminated due to futility. LY2495655 did not improve overall survival: the hazard ratio was 1.70 (90% confidence interval, 1.1-2.7) for 300 mg vs. placebo and 1.3 (0.82-2.1) for 100 mg vs. placebo (recommended doses). Progression-free survival results were consistent with the overall survival results. A numerically higher hazard ratio was observed in patients with weight loss (WL) of ≥5% (cachexia) than with
- Published
- 2018
45. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial
- Author
-
Kohei Shitara, Mustafa Özgüroğlu, Yung-Jue Bang, Maria Di Bartolomeo, Mario Mandalà, Min-Hee Ryu, Lorenzo Fornaro, Tomasz Olesiński, Christian Caglevic, Hyun C Chung, Kei Muro, Eray Goekkurt, Wasat Mansoor, Raymond S McDermott, Einat Shacham-Shmueli, Xinqun Chen, Carlos Mayo, S Peter Kang, Atsushi Ohtsu, Charles S Fuchs, Guillermo Lerzo, Juan Manuel O'Connor, Guillermo Ariel Mendez, James Lynam, Niall Tebbutt, Mark Wong, Andrew Strickland, Chris Karapetis, David Goldstein, Paul Vasey, Jean-Luc Van Laethem, Eric Van Cutsem, Scott Berry, Mark Vincent, Bettina Muller, Felipe Rey, Angela Zambrano, Joaquin Guerra, Merete Krogh, Lene Baeksgaard, Mette Yilmaz, Anneli Elme, Andrus Magi, Paivi Auvinen, Tuomo Alanko, Markus Moehler, Volker Kunzmann, Thomas Seufferlein, Peter Thuss-Patience, Thomas Hoehler, Georg Haag, Salah-Eddin Al-Batran, Hugo Castro, Karla Lopez, Mynor Aguilar Vasquez, Mario Sandoval, Ka On Lam, Sinead Cuffe, Cathy Kelly, Ravit Geva, Ayala Hubert, Alex Beny, Baruch Brenner, Aprile Giuseppe, Alfredo Falcone, Evaristo Maiello, Rodolfo Passalacqua, Vincenzo Montesarchio, Hiroki Hara, Keisho Chin, Tomohiro Nishina, Yoshito Komatsu, Nozumo Machida, Shuichi Hironaka, Taroh Satoh, Takao Tamura, Naotaoshi Sugimoto, Haruhiko Cho, Yashushi Omuro, Ken Kato, Masahiro Goto, Ichinosuke Hyodo, Kazuhiro Yoshida, Hideo Baba, Taito Esaki, Junji Furuse, Wan Zamaniah Wan Mohammed, Carlos Hernandez Hernandez, Juan Casas Garcia, Adriana Dominguez Andrade, Katriona Clarke, Geir Hjortland, Nils Glenjen, Tomasz Kubiatowski, Jassem Jacek, Marek Wojtukiewicz, Sergey Lazarev, Yuri Lancukhay, Sergey Afanasayev, Vladimir Moiseyenko, Vladimir Kostorov, Svetlana Protsenko, Vadim Shirinkin, Dina Sakaeva, Natalia Fadeeva, Wei Peng Yong, Chau Hsien Matthew Ng, Barbara Robertson, Bernardo Rapaport, Graham Cohen, Lydia Dreosti, Paul Ruff, Conrad Jacobs, Gregory Landers, Waldemar Szpak, Sang-Young Roh, Jeeyun Lee, Yeul Hong Kim, Hyun Cheol Chung, Maria Alsina Maqueda, Federico Longo Munoz, Andres Cervantes Aguilar, Enrique Aranda Aguilar, Pilar Garcia Alfonso, Fernando Rivera, Jaime Feliu Batle, Roberto Pazo Cid, Kun-Huei Yeh, Jen-Shi Chen, Yee Chao, Chia-Jui Yen, Oguz Kara, Suayib Yalcin, Daniel Hochhauser, Ian Chau, Al Benson, Veena Shankaran, Walid Shaib, Philip Philip, Vivek Sharma, Robert Siegel, Weijing Sun, Zev Wainberg, Ben George, Andrea Bullock, Samuel Myrick, Josephine Faruol, Richard Siegel, Timothy Larson, Carlos Becerra, Suresh Ratnam, Donald A. Richards, and Stephen L. Riche
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Esophageal Neoplasms ,Paclitaxel ,medicine.medical_treatment ,Population ,Phases of clinical research ,Pembrolizumab ,Adenocarcinoma ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Stomach Neoplasms ,Internal medicine ,medicine ,Humans ,Infusions, Intravenous ,education ,Survival rate ,Aged ,Neoplasm Staging ,education.field_of_study ,Chemotherapy ,business.industry ,Hazard ratio ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,Survival Rate ,Treatment Outcome ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Female ,Esophagogastric Junction ,business - Abstract
Summary Background Patients with advanced gastric or gastro-oesophageal junction cancer that progresses on chemotherapy have poor outcomes. We compared pembrolizumab with paclitaxel in patients with advanced gastric or gastro-oesophageal junction cancer that progressed on first-line chemotherapy with a platinum and fluoropyrimidine. Methods This randomised, open-label, phase 3 study was done at 148 medical centres in 30 countries. Eligible patients were randomised (1:1) in blocks of four per stratum with an interactive voice-response and integrated web-response system to receive either pembrolizumab 200 mg every 3 weeks for up to 2 years or standard-dose paclitaxel. Primary endpoints were overall survival and progression-free survival in patients with a programmed cell death ligand 1 (PD-L1) combined positive score (CPS) of 1 or higher. Safety was assessed in all patients, irrespective of CPS. The significance threshold for overall survival was p=0·0135 (one-sided). This trial is registered at ClinicalTrials.gov, number NCT02370498. Findings Between June 4, 2015, and July 26, 2016, 592 patients were enrolled. Of the 395 patients who had a PD-L1 CPS of 1 or higher, 196 patients were assigned to receive pembrolizumab and 199 patients were assigned to receive paclitaxel. As of Oct 26, 2017, 326 patients in the population with CPS of 1 or higher had died (151 [77%] of 196 patients in the pembrolizumab group and 175 [88%] of 199 patients in the paclitaxel group). Median overall survival was 9·1 months (95% CI 6·2–10·7) with pembrolizumab and 8·3 months (7·6–9·0) with paclitaxel (hazard ratio [HR] 0·82, 95% CI 0·66–1·03; one-sided p=0·0421). Median progression-free survival was 1·5 months (95% CI 1·4–2·0) with pembrolizumab and 4·1 months (3·1–4·2) with paclitaxel (HR 1·27, 95% CI 1·03–1·57). In the total population, grade 3–5 treatment-related adverse events occurred in 42 (14%) of the 294 patients treated with pembrolizumab and 96 (35%) of the 276 patients treated with paclitaxel. Interpretation Pembrolizumab did not significantly improve overall survival compared with paclitaxel as second-line therapy for advanced gastric or gastro-oesophageal junction cancer with PD-L1 CPS of 1 or higher. Pembrolizumab had a better safety profile than paclitaxel. Additional trials of pembrolizumab in gastric and gastro-oesophageal cancer are ongoing. Funding Merck Sharp & Dohme, a subsidiary of Merck & Co.
- Published
- 2018
46. Abstract P5-21-27: Efficacy and safety of ribociclib plus letrozole in US patients enrolled in the MONALEESA-2 study
- Author
-
Donald A. Richards, Lowell L. Hart, Sara M. Tolaney, M. Miller, D. A. Yardley, Paul Richards, JT Beck, Tania Small, Bhuvaneswari Ramaswamy, Gabriel N. Hortobagyi, Michaela Tsai, Francisco J. Esteva, Sibel Blau, Sami Diab, Joseph A. Sparano, Timothy J. Pluard, Das Purkayastha, Patrick S. Ward, and A.M. Favret
- Subjects
Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Letrozole ,Population ,Hazard ratio ,Neutropenia ,medicine.disease ,Interim analysis ,Placebo ,Gastroenterology ,Oncology ,Internal medicine ,medicine ,Population study ,education ,business ,Febrile neutropenia ,medicine.drug - Abstract
Background: Endocrine-based therapy is the standard of care for patients with hormone receptor–positive (HR+), human epidermal growth factor receptor 2–negative (HER2−) advanced breast cancer (ABC), in whom delaying disease progression is the primary goal. In the randomized Phase 3 MONALEESA-2 study (ClinicalTrials.gov identifier, NCT01958021), first-line ribociclib (a selective inhibitor of cyclin-dependent kinases 4 and 6) + letrozole significantly prolonged progression-free survival (PFS) compared with placebo + letrozole in postmenopausal women with HR+, HER2− ABC (hazard ratio, 0.556; 95% confidence interval [CI], 0.429–0.720) at the interim analysis cutoff date. Here, we present safety and efficacy data from US patients who were enrolled in the MONALEESA-2 study. Methods: Postmenopausal women (N=668) with HR+, HER2− ABC who did not receive prior systemic treatment for ABC and had an Eastern Cooperative Oncology Group performance status score of ≤1, adequate bone marrow and organ function, and no history of active cardiac dysfunction were randomized 1:1 to receive either ribociclib (600 mg/d, 3 weeks on/1 week off) + letrozole (2.5 mg/d, continuous) or placebo + letrozole. The primary end point was locally assessed PFS. The data cutoff for this analysis was January 29, 2016. Results: Baseline demographics, patient disposition, and prior therapy were well balanced across treatment groups in the US subset (N=213; ribociclib group, n=100; placebo group, n=113). Median treatment duration was 16.3 months. In US patients, median PFS was significantly prolonged in the ribociclib group (median PFS, not reached [NR]; 95% CI, 17.1 months to NR) versus the placebo group (median PFS, 14.4 months [95% CI, 11.1–18.4 months]; hazard ratio, 0.451 [95% CI, 0.281–0.724]; P=0.000363), with a median follow-up of 11.0 months. In a subsequent preplanned analysis of overall survival, median PFS was 27.6 months in the ribociclib group versus 15.0 months in the placebo group (hazard ratio, 0.527; 95% CI, 0.351–0.793). The overall response rate (ORR) was significantly greater in the ribociclib group (ORR, 38.0%; 95% CI, 28.5–47.5%) versus the placebo group (ORR, 26.5%; 95% CI, 18.4–34.7%; P=0.040). A trend toward benefit was observed with ribociclib in all subgroups (including age, disease burden, prior therapy, and biomarker status), which was consistent with the overall population. No on-treatment deaths occurred. The most common all-grade adverse events, irrespective of causality, were neutropenia (ribociclib, 69.0%; placebo, 3.7%), nausea (ribociclib, 65.0%; placebo, 39.4%), fatigue (ribociclib, 56.0%; placebo, 46.8%), and diarrhea (ribociclib, 41.0%; placebo, 32.1%). The most common grade 3 (≥25% in either group) and grade 4 (≥10% in either group) AE was neutropenia (grade 3: ribociclib, 39.0%; placebo, 0%; grade 4: ribociclib, 14.0%; placebo, 0%). Two patients (2.0%) in the ribociclib group and none in the placebo group experienced febrile neutropenia. Conclusions: The US patients in the MONALEESA-2 study derived significant PFS benefit from ribociclib + letrozole compared with placebo + letrozole, with a 55% reduction in the relative risk of disease progression. The safety profile was consistent with and comparable to the global study population. Citation Format: Yardley DA, Hart L, Favret A, Blau S, Diab S, Richards D, Sparano J, Beck JT, Richards P, Ward P, Ramaswamy B, Tsai M, Pluard T, Tolaney S, Esteva F, Small T, Purkayastha D, Miller M, Hortobagyi G. Efficacy and safety of ribociclib plus letrozole in US patients enrolled in the MONALEESA-2 study [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P5-21-27.
- Published
- 2018
47. Abstract PD5-08: Not presented
- Author
-
Richard M. Elledge, Donald A. Richards, Sharon Wilks, Peter Kabos, Virginia G. Kaklamani, H Jiang, D Wang, Aditya Bardia, A O'Neill, Wael A. Harb, and F Garner
- Subjects
Cancer Research ,Oncology - Abstract
This abstract was not presented at the symposium.
- Published
- 2018
48. Whole blood FPR1 mRNA expression predicts both non-small cell and small cell lung cancer
- Author
-
Kirk Ryden, D. K. Hogarth, David Mallery, Glen J. Weiss, Donald A. Richards, Scott Morris, Troy Shelton, Harvey I. Pass, Anil Vachani, William N. Rom, and George C. Runger
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,TaqMan ,Medicine ,Biomarker (medicine) ,Blood test ,Non small cell ,business ,Lung cancer ,Lung cancer screening ,Whole blood - Abstract
While long-term survival rates for early-stage lung cancer are high, most cases are diagnosed in later stages that can negatively impact survival rates. We aim to design a simple, single biomarker blood test for early-stage lung cancer that is robust to preclinical variables and can be readily implemented in the clinic. Whole blood was collected in PAXgene tubes from a training set of 29 patients, and a validation set of 260 patients, of which samples from 58 patients were prospectively collected in a clinical trial specifically for our study. After RNA was extracted, the expressions of FPR1 and a reference gene were quantified by an automated one-step Taqman RT-PCR assay. Elevated levels of FPR1 mRNA in whole blood predicted lung cancer status with a sensitivity of 55% and a specificity of 87% on all validation specimens. The prospectively collected specimens had a significantly higher 68% sensitivity and 89% specificity. Results from patients with benign nodules were similar to healthy volunteers. No meaningful correlation was present between our test results and any clinical characteristic other than lung cancer diagnosis. FPR1 mRNA levels in whole blood can predict the presence of lung cancer. Using this as a reflex test for positive lung cancer screening computed tomography scans has the potential to increase the positive predictive value. This marker can be easily measured in an automated process utilizing off-the-shelf equipment and reagents. Further work is justified to explain the source of this biomarker.
- Published
- 2018
49. 1191O MRTX-500: Phase II trial of sitravatinib (sitra) + nivolumab (nivo) in patients (pts) with non-squamous (NSQ) non-small cell lung cancer (NSCLC) progressing on or after prior checkpoint inhibitor (CPI) therapy
- Author
-
Manish R. Patel, Collin M. Blakely, Igor I. Rybkin, D. Bruno, D.M. Waterhouse, Kai He, D. Hong, A. Pham, Edward B. Garon, David Berz, Ticiana A. Leal, Donald A. Richards, Alexander I. Spira, Robert M. Jotte, Wade T. Iams, R. Shazer, L. Latven, and X. Yan
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immune checkpoint inhibitors ,non-small cell lung cancer (NSCLC) ,Hematology ,medicine.disease ,Non squamous ,Sitravatinib ,Internal medicine ,medicine ,In patient ,Nivolumab ,business - Published
- 2021
50. 1123O Evaluation of cell-free DNA approaches for multi-cancer early detection
- Author
-
Arash Jamshidi, John F. Beausang, Alan H. Bryce, Michael V. Seiden, M.K. Tummala, Charles Swanton, Kathryn N. Kurtzman, Kristi McIntyre, Donald A. Richards, Earl Hubbell, T.J. Yeatman, Minetta C. Liu, Oliver Venn, David D. Thiel, Mikkael A. Sekeres, Eric A. Klein, N. Zhang, Allen Lee Cohn, and Chenlu Hou
- Subjects
Oncology ,Cell-free fetal DNA ,business.industry ,Cancer research ,Medicine ,Hematology ,business ,Cancer Early Detection - Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.