35 results on '"J. Garcia-Corbacho"'
Search Results
2. Author Correction: Results of the phase IIa RADICAL trial of the FGFR inhibitor AZD4547 in endocrine resistant breast cancer
- Author
-
R. C. Coombes, P. D. Badman, J. P. Lozano-Kuehne, X. Liu, I. R. Macpherson, I. Zubairi, R. D. Baird, N. Rosenfeld, J. Garcia-Corbacho, N. Cresti, R. Plummer, A. Armstrong, R. Allerton, D. Landers, H. Nicholas, L. McLellan, A. Lim, F. Mouliere, O. E. Pardo, V. Ferguson, and M. J. Seckl
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
3. 125P Assessing the accuracy of multiple prognostic scores for immune checkpoint inhibitors (ICI) in patients with advanced solid tumors
- Author
-
J. Garcia-Corbacho, A. Indacochea, A. Gonzalez, I. Victoria Ruiz, D. Moreno Fernández, D.S. Pesantez Coronel, L. Angelats, A. Modrego, E. Sanfeliu Torres, O. Castillo, P. Blasco, L. Mezquita, N. Vinolas Segarra, M. Nogue Aliguer, B. Adamo, N. Baste Rotllan, T. Sauri Nadal, M. Juan, A. Prat, and F. Schettini
- Subjects
Oncology ,Hematology - Published
- 2022
- Full Text
- View/download PDF
4. 44P Biomarker analysis from a phase Ib, multicenter, open-label clinical trial of talimogene laherparepvec (T-VEC) injected (inj) into metastatic and primary liver tumors
- Author
-
J.R. Hecht, M. Peeters, M. Martin Jimenez, M. Pless, A. Cubillo, J. Garcia-Corbacho, N. Mach, A. Digklia, H. Park, V. Subbiah, K. Gorski, A. Anderson, C. Liu, W. Snyder, and J. Chesney
- Subjects
Oncology ,Hematology - Published
- 2021
- Full Text
- View/download PDF
5. P86.03 A Phase 2 Study of Erdafitinib in Patients with Advanced Solid Tumors and Fibroblast Growth Factor Receptor Gene Alterations
- Author
-
Y.Y. Lau, J. Garcia-Corbacho, J. Tabernero, C. Hammond, Christophe Massard, D. Hargrave, S. Little, Toshihiko Doi, A. Santiago-Walker, Shukui Qin, Martin Schuler, J. Tolbert, Olaf Witt, C. Lu-Emerson, C. Moy, David M. Hyman, Hussein Sweiti, and Shubham Pant
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,Erdafitinib ,business.industry ,Fibroblast growth factor receptor ,Cancer research ,Medicine ,Phases of clinical research ,In patient ,business ,Gene - Published
- 2021
- Full Text
- View/download PDF
6. CX-2009, a CD166-directed probody drug conjugate (PDC): Results from the first-in-human study in patients (Pts) with advanced cancer including breast cancer (BC)
- Author
-
Funda Meric-Bernstam, Howard A. Burris, Rachel E. Sanborn, Patricia LoRusso, James J. Harding, Amy Weise, Lee S. Rosen, Bert H. O'Neil, Randy F. Sweis, Rachel Li, Iván Victoria, Hendrik-Tobias Arkenau, Joyce F. Liu, Mary J. Fidler, Nataliya Volodymyrivna Uboha, J. Garcia-Corbacho, Mark Stroh, John W. Frye, Alexander I. Spira, and Valentina Boni
- Subjects
Drug ,Cancer Research ,business.industry ,media_common.quotation_subject ,First in human ,medicine.disease ,Advanced cancer ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Microtubule Inhibitor ,Medicine ,In patient ,business ,ALCAM ,030215 immunology ,media_common ,Conjugate - Abstract
526 Background: CX-2009 is a PROBODY drug conjugate (PDC) directed against CD166 (ALCAM) and conjugated to DM4, a potent microtubule inhibitor (MTI). CD166 is overexpressed in carcinomas but is also ubiquitously expressed in normal epithelium and thus has not been previously considered a viable target for a traditional antibody drug conjugate. PDCs have a peptide mask that blocks normal tissue binding and can be removed by tumor-associated proteases, thereby limiting off-tumor/on-target binding. CX-2009 demonstrated preclinical activity in multiple solid tumor models. Here we report results of the first in human study in patients with advanced cancer. Methods: In this phase I multi-part dose-escalation study, pts with advanced solid tumors received CX-2009 0.25–10 mpk IV every 14 or 21 days (Q2W or Q3W). Tumor types were selected based on expected high CD166 expression and MTI sensitivity. Results: The dose-escalation phase of the trial enrolled 43 pts; 49 additional pts were subsequently enrolled between 4–10 mpk to collect biomarker data and define the recommended phase II dose (RP2D), for a total of 92 pts as of 30 Nov 2019 (39 pts with breast cancer [BC], 22 ovarian [OC], 12 non-small cell lung [NSCLC], 9 head/neck squamous cell [HNSCC], 10 other) with a median of 6 (range 1–19) prior therapies. Median number of CX-2009 doses was 2 (range, 1–15). For Q3W dosing, one dose limiting toxicity (DLT; grade 3 vomiting) was observed at 8 mpk; MTD was not reached up to 10 mpk. The RP2D for Q3W schedule was 7 mpk based on safety, dose-response, and population pharmacokinetic simulations. Q2W dosing continues; DLTs were observed at 6 mpk. Common treatment-related adverse events (TRAEs) at 7 mpk (n=9) were nausea (44%), fatigue, infusion-related reactions (both 33%), vomiting and arthralgias (both 22%). Grade 3 TRAEs occurred in 2 pts (nausea/vomiting; peripheral neuropathy). No pts discontinued at 7 mpk due to TRAEs. Ocular toxicity was dose dependent; mild to moderate reversible keratitis/blurred vision was seen in 3 pts at 7 mpk and mitigated by ocular prophylaxis. Partial responses were seen in 8 pts (2 confirmed, both HR+/HER2- BC) treated between 4–10 mpk, including BC (n=5), OC (n=2), and HNSCC (n=1). SD (≥1 on-study scan) was observed in 21 pts, 5 had SD ≥3 mos. Conclusions: CX-2009 at 7 mpk is the RP2D on Q3W schedule. Phase II expansion has begun in pts with HR+/HER2- BC. The Q2W schedule will continue to enroll pts to define the RP2D. CX-2009 will also be studied in combination with CX-072, a PD-L1 PROBODY therapeutic ( NCT03149549 ) Clinical trial information: NCT03149549 .
- Published
- 2020
- Full Text
- View/download PDF
7. An Extended Phase Ib Study of Epertinib, an Orally Active Reversible Dual EGFR/HER2 Tyrosine Kinase Inhibitor, in Patients with Solid Tumours
- Author
-
J Posner, Antoine Italiano, J Garcia-Corbacho, H-T. Arkenau, D Tosi, Richard H. Wilson, Antoine Adenis, Michael Flynn, S Deva, A Arimura, Gabriel Mak, Maud Toulmonde, Ken Donaldson, I Kawabata, Ruth Plummer, Richard D. Baird, Martin Forster, James Spicer, Institut Bergonié [Bordeaux], UNICANCER, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Centre Régional de Lutte contre le Cancer Oscar Lambret [Lille] (UNICANCER/Lille), Université Lille Nord de France (COMUE)-UNICANCER, Université de Lille-UNICANCER, Baird, Richard [0000-0001-7071-6483], and Apollo - University of Cambridge Repository
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Loperamide ,medicine.drug_class ,EGFR ,Tyrosine kinase inhibitor ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Epertinib ,Gastroenterology ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Pharmacokinetics ,Neoplasms ,Internal medicine ,HER2 ,Humans ,Medicine ,In patient ,030212 general & internal medicine ,Dosing ,Adverse effect ,skin and connective tissue diseases ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,business.industry ,Middle Aged ,medicine.disease ,3. Good health ,Treatment Outcome ,S-222611 ,Oncology ,Tolerability ,030220 oncology & carcinogenesis ,Quinazolines ,Female ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,medicine.drug - Abstract
Background:\ud Dose-escalation of epertinib (S-222611), a new potent oral EGFR/HER2 inhibitor, has established a recommended daily dose of 800 mg in patients with solid tumours. In this study, we have recruited a larger number of patients to assess further the safety, tolerability, pharmacokinetics (PKs) and antitumour activity.\ud \ud Patients and Methods:\ud Patients with solid tumours expressing EGFR or HER2 received a single dose of epertinib at 800 mg on Day 1 to assess PK over 7 days, followed by continuous once-daily dosing from Day 8.\ud \ud Results:\ud We treated 76 patients with breast (n = 27), upper gastrointestinal (GI; n = 30), head and neck (n = 12) or renal cancers (n = 7). Epertinib was well-tolerated with mostly grade I and II adverse events (AEs). The most frequent AE was diarrhoea, which was generally manageable with loperamide. The objective response rate (ORR) in patients with heavily pretreated breast and upper GI cancers was 16.0% (4 PRs) and 8.3% (1CR, 1PR), respectively. All six responding patients had HER2-positive tumours; the ORR for HER2-positive breast and upper GI cancer populations was 19.0% and 20.0%. Partial response in the brain disease of one breast cancer patient lasted 7.5 months.\ud \ud Conclusion:\ud Once-daily dosing of epertinib at 800 mg was well-tolerated and demonstrated promising antitumour activity in patients with heavily pretreated HER2-positive breast and upper GI cancer, including those with brain metastases.\ud \ud EudraCT number:\ud 2009-017817-31.
- Published
- 2018
- Full Text
- View/download PDF
8. Preliminary results of the first-in-human, dose-finding PROCLAIM-CX-072 trial evaluating the PD-L1 probody therapeutic CX-072 in combination with ipilimumab (ipi) in patients (pts) with advanced solid tumors
- Author
-
Mary J. Fidler, Ruth Plummer, Karen A. Autio, E. G. E. de Vries, Rachel E. Sanborn, H.-T. Arkenau, C. W. Menke-van der Houven van Oordt, Anthony B. El-Khoueiry, Valentina Boni, Jerzy Wydmański, Patricia LoRusso, R. Humphrey, J. Garcia-Corbacho, Nataliya Volodymyrivna Uboha, Fiona C Thistlethwaite, Matthias Will, Medical oncology, CCA - Imaging and biomarkers, and CCA - Treatment and quality of life
- Subjects
0301 basic medicine ,Oncology ,Brachial Plexus Neuritis ,medicine.medical_specialty ,biology ,business.industry ,Ipilimumab ,Hematology ,First in human ,03 medical and health sciences ,Dose finding ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,PD-L1 ,medicine ,biology.protein ,In patient ,business ,medicine.drug - Published
- 2018
- Full Text
- View/download PDF
9. Abstract P4-14-26: Phase I expansion of S-222611, a reversible inhibitor of EGFR and HER2, in advanced solid tumors, including HER2-positive breast cancer patients with brain metastases
- Author
-
Linda Hogarth, Debashis Sarker, Antoine Italiano, J Posner, N Cresti, Duncan I. Jodrell, K Donaldson, K Kawaguchi, S Deva, J. Garcia-Corbacho, Richard D. Baird, Ruth Plummer, A Arimura, Eugene P. Frenkel, James Spicer, and H.-T. Arkenau
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,medicine.drug_class ,Population ,Lapatinib ,Tyrosine-kinase inhibitor ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,skin and connective tissue diseases ,Adverse effect ,education ,education.field_of_study ,business.industry ,Cancer ,medicine.disease ,Metastatic breast cancer ,030104 developmental biology ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
BACKGROUND S-222611 is an oral, reversible ErbB tyrosine kinase inhibitor of EGFR and HER2 with potent pre-clinical activity. MTD was not reached during the dose-escalation phase, (maximum dose 1600 mg QD). PK and efficacy data supported a daily dose of 800 mg. An expansion cohort of patients has been treated to further explore safety and efficacy. METHODS Subjects with advanced solid tumors expressing EGFR and/or overexpressing HER2 were enrolled. S-222611 800 mg daily was administered until disease progression or unacceptable toxicity. RESULTS 76 patients were included in this phase 1 expansion cohort with a variety of tumor types. Dose reduction was required because of adverse events in 15 patients; the most frequent of which being diarrhea and elevated bilirubin. Two patients discontinued treatment due to drug- related adverse events. Of the 25 patients with HER2-positive metastatic breast cancer (MBC), 4 partial responses were observed, and prolonged stable disease (≥ 6 months) was observed in 3 additional patients. These 25 patients had received prior HER2-directed therapy as shown in Table 1. Table 1. Prior therapies received by patients with HER2-positive MBCPrior therapyn (%)Trastuzumab22 (88)T-DM13 (12)Lapatinib16 (64)Chemotherapy23 (92) Six of these patients had brain metastases, in whom 1 intracranial response and 2 prolonged stable disease (≥ 6 months) were observed (Table 2). Table 2. HER2-positive MBC patients with brain metastases - best overall response to S-222611Pts #HER2 IHCBrain metastasesBest overall response (RECIST 1.1)Patient 13+Target lesionPRPatient 23+Target lesionSD (≥12 M)Patient 33+Target lesionSD (6.0 M)Patient 43+Non-target lesionSD (4.7 M)Patient 53+Non-target lesionSD (3.3 M)Patient 63+Non-target lesionNE The patient showing intracranial response was previously treated with lapatinib and capecitabine after diagnosis of BM. CONCLUSIONS S-222611 was well tolerated at a dose of 800 mg once daily. Anti-tumour activity, including shrinkage of brain metastases, was evident in a heavily pre-treated population of patients with HER2-positive breast cancer. Citation Format: Baird RD, Arkenau H-T, Deva S, Cresti N, Garcia-Corbacho J, Hogarth L, Frenkel E, Kawaguchi K, Arimura A, Donaldson K, Posner J, Sarker D, Jodrell D, Plummer R, Spicer J, Italiano A. Phase I expansion of S-222611, a reversible inhibitor of EGFR and HER2, in advanced solid tumors, including HER2-positive breast cancer patients with brain metastases. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P4-14-26.
- Published
- 2016
- Full Text
- View/download PDF
10. Phase 1 dose-escalation study of S-222611, an oral reversible dual tyrosine kinase inhibitor of EGFR and HER2, in patients with solid tumours
- Author
-
Ruth Plummer, Aneta Suder, N Cresti, J. Garcia Corbacho, I Kawabata, Duncan I. Jodrell, Linda Hogarth, James Spicer, S. Matsumoto, Eugene P. Frenkel, K Donaldson, J Posner, Debashis Sarker, Richard D. Baird, Baird, Richard [0000-0001-7071-6483], Jodrell, Duncan [0000-0001-9360-1670], and Apollo - University of Cambridge Repository
- Subjects
Adult ,Diarrhea ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Metabolic Clearance Rate ,Receptor, ErbB-2 ,Nausea ,medicine.drug_class ,EGFR ,Administration, Oral ,Phases of clinical research ,Pharmacology ,Lapatinib ,Drug Administration Schedule ,Tyrosine-kinase inhibitor ,Young Adult ,Pharmacokinetics ,Trastuzumab ,HER2 ,Neoplasms ,Internal medicine ,medicine ,Humans ,HER4 ,Protein Kinase Inhibitors ,Fatigue ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Standard treatment ,Phase 1 clinical trial ,Exanthema ,Middle Aged ,Rash ,ErbB Receptors ,Treatment Outcome ,Area Under Curve ,Protein kinase inhibitor ,Quinazolines ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
Background S-222611 is a reversible inhibitor of EGFR, HER2 and HER4 with preclinical activity in models expressing these proteins. We have performed a Phase 1 study to determine safety, maximum tolerated dose (MTD), pharmacokinetic profile (PK) and efficacy in patients with solid tumours expressing EGFR or HER2. Patients and methods Subjects had advanced tumours not suitable for standard treatment, expressing EGFR or HER2, and/or with amplified HER2 . Daily oral doses of S-222611 were escalated from 100 mg to 1600 mg. Full plasma concentration profiles for drug and metabolites were obtained. Results 33 patients received S-222611. It was well tolerated, and the most common toxicities, almost all mild (grade 1 or 2), were diarrhoea, fatigue, rash and nausea. Only two dose-limiting toxicities occurred (diarrhoea and rash), which resolved on interruption. MTD was not reached. Plasma exposure increased with dose up to 800 mg, exceeding levels eliciting pre-clinical responses. The plasma terminal half-life was more than 24 h, supporting once daily dosing. Responses were seen over a wide range of doses in oesophageal, breast and renal tumours, including a complete clinical response in a patient with HER2-positive breast carcinoma previously treated with lapatinib and trastuzumab. Four patients have remained on treatment for more than 12 months. Downregulation of pHER3 was seen in paired tumour biopsies from a responding patient. Conclusions Continuous daily oral S-222611 is well tolerated, modulates oncogenic signalling, and has significant antitumour activity. The recommended Phase 2 dose, based on PK and efficacy, is 800 mg/day.
- Published
- 2015
- Full Text
- View/download PDF
11. Abstract P4-12-24: Phase I trial of S-222611, a dual tyrosine kinase inhibitor of EGFR and HER2, with preliminary evidence of efficacy in patients (pts) with heavily-pretreated HER2-positive metastatic breast cancer
- Author
-
F Gao, J. Garcia Corbacho, Richard D. Baird, Ruth Plummer, Emma Beddowes, H.-T. Arkenau, Duncan I. Jodrell, C. Lemech, I Kawabata, M Saggese, Martin Forster, K Donaldson, J Posner, N Cresti, M Flynn, and James Spicer
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.drug_class ,Cancer ,Pharmacology ,Lapatinib ,medicine.disease ,Rash ,Metastatic breast cancer ,Gastroenterology ,Tyrosine-kinase inhibitor ,Oncology ,Pharmacokinetics ,Trastuzumab ,Pharmacodynamics ,Internal medicine ,Medicine ,medicine.symptom ,business ,medicine.drug - Abstract
Background and rationale: S-222611 is a novel, oral, reversible inhibitor of EGFR, HER2 and HER4 with an improved preclinical profile compared with first-generation pan-HER inhibitors. Research objectives: We conducted a phase I study to determine safety, maximum tolerated dose (MTD), pharmacokinetics (PK), pharmacodynamics (PD) and preliminary efficacy in pts with solid tumors expressing EGFR or HER2. Patients and methods: Pts with advanced solid tumors and biopsies confirming EGFR and/or HER2 expression were treated with continuous daily oral doses of S-222611. The dose of S-222611 was escalated in cohorts of at least 3 pts from 100mg to 1600mg. PK profiles were obtained in all pts, and PD assays for pEGFR, pHER2 and pHER3 were performed on paired tumor biopsies in selected pts. Results: A total of 50 pts have been treated to-date; age 25-80y; 20 female; 13 with HER2-positive metastatic breast cancer (HER2+ MBC). S-222611 was generally well tolerated with two dose-limiting toxicities in the dose-escalation phase: rash at 1200mg; diarrhea at 1600mg. MTD was not defined. Diarrhea was the most frequent toxicity, but was rarely worse than grade 1/2. Nausea, rash, anorexia and fatigue were also seen. Bilirubin rises with normal transaminases were observed. Plasma concentrations of S-222611 increased with dose up to 800mg, which was the dose selected for the expansion phase. Steady state values of Cmax and AUC0-24 at this dose were in the effective range of concentrations in mouse models. Average t½ of 33h was consistent with once daily dosing. Tumor responses were seen over the full dose range tested (100-1600mg), with four pts on treatment for >12 months. Out of 13 HER2+ MBC pts, one clinical complete response (CR) was observed for >12 months, and four partial responses (PRs) were also seen (2 confirmed, 2 unconfirmed), yielding an overall response rate of 38% in this small group (Table 1). All 5 responding patients had previously progressed on trastuzumab (T); 4 of them had also progressed on lapatinib (L); and one patient had progressed on T, L and T-DM1. Out of 6 breast patients with PD results available, the 3 pts with a decrease in pHER3 all responded, whereas the 3 pts with no change or increase in pHER3 all had disease progression. Conclusion: S-222611 was well tolerated in doses up to 1600mg daily with diarrhea, nausea and rash being readily manageable. Significant antitumor activity has been observed in patients with heavily pre-treated HER2+ MBC, including those progressing on prior T, L and T-DM1. Rates of grade 3 diarrhea appear markedly lower than those reported for other second-generation pan-HER TKIs. The recommended phase 2 dose, based on PK and clinical activity, is 800mg/day. Table 1. HER2+ MBC pts with tumor shrinkage after S-222611 treatmentPatient numberPrior progression on trastuzumab-based regimenPrior progression on lapatinib-based regimenBest responseMonths on treatment (at time of data lock)S01014YYclinical CR20.3m (ongoing)S03034YNconfirmed PR11.5m (ongoing)S04046YYconfirmed PR6.0m (ongoing)S01030YYunconfirmed PRwithdrawn at 3.7m with progressionS01047YYunconfirmed PR4.8m (ongoing) Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P4-12-24.
- Published
- 2013
- Full Text
- View/download PDF
12. The first-in-human, dose-finding PROCLAIM-CX-072 trial to assess the antitumor activity and tolerability of the probody therapeutic CX-072 as monotherapy and in combination with ipilimumab or vemurafenib in solid advanced tumors and lymphomas
- Author
-
Valentina Boni, Jaime Feliu, B. Irving, E.G.E. de Vries, Matthias Will, Z. Horvath, Jerzy Wydmański, J. Garcia-Corbacho, Fiona C Thistlethwaite, I. Bondarenko, and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
Antitumor activity ,Dose finding ,Oncology ,Tolerability ,business.industry ,Medicine ,Ipilimumab ,Hematology ,First in human ,Pharmacology ,business ,Vemurafenib ,medicine.drug - Published
- 2017
- Full Text
- View/download PDF
13. AVEVAC: A phase I-II trial with avelumab plus autologous dendritic cell (ADC) vaccine in pre-treated mismatch repair-proficient (MSS) metastatic colorectal cancer (mCRC) patients (GEMCAD 16-02)
- Author
-
J. Garcia-Corbacho, M. Español Rego, R. Cabezón, Jorge Aparicio, Miquel Lozano, Carlos Fernández-Martos, Juan Maurel, E. Elez Fernandez, A. Ruiz Casado, Jessica Cid, X. García de Albéniz, Nohra Cecilia Rodríguez, D. Benitez Ribas, Pilar Escudero, A. Ginés, V. Alonso, and L. Bianchi
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,Hematology ,Dendritic cell ,medicine.disease ,Avelumab ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Phase i ii ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,DNA mismatch repair ,business ,medicine.drug - Published
- 2018
- Full Text
- View/download PDF
14. Preliminary results of PROCLAIM-CX-072: The first-in-human, dose-finding trial of PD-L1 probody therapeutic CX-072 as monotherapy in patients (pts) with advanced solid tumors
- Author
-
Nataliya Volodymyrivna Uboha, E.G.E. de Vries, Karen A. Autio, Matthias Will, Fiona C Thistlethwaite, Daniel C. Cho, H.-T. Arkenau, Patrick A. Ott, R. Humphrey, J. Garcia-Corbacho, Aung Naing, Alexander I. Spira, and Valentina Boni
- Subjects
0301 basic medicine ,Brachial Plexus Neuritis ,Solid tumour ,medicine.medical_specialty ,biology ,business.industry ,Urology ,Hematology ,First in human ,03 medical and health sciences ,Dose finding ,030104 developmental biology ,Oncology ,PD-L1 ,biology.protein ,Medicine ,In patient ,business - Published
- 2018
- Full Text
- View/download PDF
15. PROCLAIM-CX-2009: A first-in-human trial to evaluate CX-2009 in adults with metastatic or locally advanced unresectable solid tumors
- Author
-
A. Yang Weaver, J. Garcia-Corbacho, James J. Harding, Mark R. Middleton, Jaime Feliu, Funda Meric-Bernstam, H. A. Burris, V. Boni, Alexander I. Spira, Matthias Will, and Volker Heinemann
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Locally advanced ,Hematology ,First in human ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business - Published
- 2017
- Full Text
- View/download PDF
16. RADICAL trial: A phase Ib/IIa study to assess the safety and efficacy of AZD4547 in combination with either anastrozole or letrozole in ER positive breast cancer patients progressing on these aromatase inhibitors (AIs)
- Author
-
Iain R. Macpherson, Donal Landers, Adrian Lim, Elizabeth Ruth Plummer, J. Garcia-Corbacho, Nicola Cresti, Xinxue Liu, Richard D. Baird, Rozenn Allerton, Ishtiaq Husain Zubairi, Charles Coombes, Hanna Nicholas, Lyndall McLellan, Philip Badman, Michael J. Seckl, and Anne C Armstrong
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Letrozole ,Disease progression ,Estrogen receptor ,Anastrozole ,Surgery ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Clinical endpoint ,biology.protein ,Breast cancer cells ,Aromatase ,business ,medicine.drug - Abstract
1059 Background: Patients with metastatic ER positive breast cancer invariably experience disease progression whilst taking AIs. Fibroblast growth factor receptor inhibitors (FGFRI) such as AZD4547 can reverse endocrine resistance in breast cancer cells. Consequently, we designed the RADICAL trial to test the safety and efficacy of AZD4547 combined with letrozole (L) or anastrozole (A). Methods: Patients with prior disease progression on either AI were initially recruited to a Phase Ib study which showed that L 2.5mg or A 1mg daily continuously could be safely combined with AZD4547 80mg twice daily on a 1wk on/1 wk off schedule. Pharmacokinetic data showed no significant interactions. Subsequently, 52 patients progressing on these AIs were recruited, either continuing, or, if other therapies had subsequently been given, restarting their prior AI together with AZD4547. Primary endpoint was change in tumour size (RECIST v 1.1) at 12 weeks compared to baseline. Results: Enrolled patients had previously received a median of 4 (range: 1-11) systemic therapies, including endocrine treatments with a median of 2 (range: 1-6). The mean tumour size change at 12 and 28 weeks was 7% (95%CI: -4%, 17%) and 8% (95%CI: -4%, 20%), respectively. Clinical benefit assessed by partial response (PR) or stable disease (SD) occurred in 36.5% (1 PR and 18 SD) and 25% (2 PR and 11 SD) of patients at 12 and 28 weeks, respectively. The median progression free survival was 3.1 months (95%CI: 2.4-5.4). Most adverse events (AEs) were G1/2 (95.3%). 11 (21%) patients developed asymptomatic AZD4547-induced retinal pigment epithelial detachment, all resolved and 1 and 6 were able to continue on study medication at full and half dose, respectively. Among 34 G3/4 AEs, only 6 were probably/possibly related to AZD4547. Out of 13 unrelated serious AEs, 2 were fatal. Conclusions: Combined AZD4547 with L or A appears to be safe and shows anti-tumour activity in advanced ER+ patients resistant to these AIs. Development of a biomarker to select patients for this therapy will facilitate future studies. Clinical trial information: NCT01791985.
- Published
- 2017
- Full Text
- View/download PDF
17. CamBMT1: A proof-of-principle phase 1b / randomised phase 2 study of afatinib penetration into brain metastases (mets) for patients undergoing neurosurgical resection, both with and without prior low-dose, targeted radiotherapy
- Author
-
A Jonson, Gillian A Whitfield, J Garcia-Corbacho, Simon Pacey, Rajesh Jena, D. Smith, Duncan I. Jodrell, Stephen J. Price, Saif S Ahmad, Martin J. Graves, Tomasz Matys, Wendi Qian, Sarah Jefferies, Richard D. Baird, Sudhir Kumar, Carlos Caldas, Anthony J. Chalmers, Colin Watts, Constanza Linossi, and Nicola Ramenatte
- Subjects
medicine.medical_specialty ,business.industry ,Targeted Radiotherapy ,Afatinib ,Low dose ,Phases of clinical research ,Hematology ,Penetration (firestop) ,Surgery ,Resection ,Oncology ,medicine ,Radiology ,business ,medicine.drug - Published
- 2016
- Full Text
- View/download PDF
18. 7129 POSTER Adjuvant Chemotherapy for High-risk Patients With Urothelial Carcinoma
- Author
-
O. Castillo Fernandez, Cristina Suarez, Xavier Maldonado, Claudia Valverde, J. Garcia-Corbacho, Joan Carles, I. Nunez, R. Morales Barrera, César Serrano, and C. Raventós
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,High risk patients ,Adjuvant chemotherapy ,business.industry ,Internal medicine ,medicine ,business ,Urothelial carcinoma - Published
- 2011
- Full Text
- View/download PDF
19. PROCLAIM-CX-072: Analysis of patients with advanced solid tumors receiving long-term treatment with CX-072, a PD-L1 probody therapeutic, as a single agent or in combination with ipilimumab
- Author
-
Ignacio Melero, Hendrik-Tobias Arkenau, Rachel E. Sanborn, Marta Gil-Martin, Nataliya Volodymyrivna Uboha, Elisabeth G.E. de Vries, Javier Lavernia, William J. Garner, Johanna C. Bendell, Iván Victoria, Aung Naing, Patricia LoRusso, Elizabeth Ruth Plummer, J. Garcia-Corbacho, Fiona C Thistlethwaite, Alexander I. Spira, Manreet Randhawa, Ferry A.L.M. Eskens, Igor Bondarenko, and Daniel C. Cho
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Long term treatment ,biology ,business.industry ,Immune checkpoint inhibitors ,Ipilimumab ,Internal medicine ,PD-L1 ,Toxicity ,medicine ,biology.protein ,Single agent ,business ,medicine.drug - Abstract
3005 Background: Monotherapy with immune checkpoint inhibitors (ICIs) has demonstrated efficacy in many cancers. Combining ICIs PD-L1 + CTLA-4 enhanced efficacy but worsened toxicity vs monotherapy; therefore, CTLA-4 dose modifications are often needed, despite a dose-response effect having been shown for efficacy. CX-072 is an investigational PD-L1 PROBODY therapeutic that is preferentially activated in the tumor microenvironment (TME); localized activation may reduce immune-related AEs (irAEs). PROCLAIM-CX-072-001 identified 10 mg/kg Q2W (Mono10) as the recommended monotherapy dose. Here we provide data for Mono10 and for dose escalation of CX-072 in combination with IPI (Combo), with a focus on long-term (≥6 mo) therapy. Methods: Mono10 was evaluated in multiple tumor types. Combo doses evaluated were CX-072 0.3–10 mg/kg and IPI 3–10 mg/kg Q3W. Patients (pts) with ≥6 mo treatment duration (≥6M-TD) were compared to those with < 6 mo of treatment ( < 6M-TD) as of November 30, 2019. Results: Disease control rates (DCR = CR+PR+SD) were 41% for Mono10 (n = 47 of 114; 10 PRs) and 37% for Combo (n = 10 of 27; 1CR + 4 PRs (1CR and 3PRs at 3 mg/kg IPI [IPI3]). Additional results are shown in the table. No treatment-related adverse events (TRAEs) led to death. The most common reason for discontinuation (dc) in all groups was disease progression. Conclusions: CX-072 monotherapy demonstrated durable responses consistent with activation of the PROBODY therapeutic in the TME. The safety profile supports the tolerability of CX-072 as monotherapy and when combined with IPI3. CX-072 + IPI3 demonstrated activity in heavily pretreated pts with various tumors. The safety profile of the combination of CX-072 with IPI3 compares favorably to historical data (grade ≥3 TRAEs 55% and leading to dc in 36%; Larkin J, et al. N Engl J Med. 2015;373:23-34). CX-072 + IPI3 is being explored in a phase 2 study in 2L melanoma Clinical trial information: NCT03993379 . [Table: see text]
20. Imlunestrant, an oral selective estrogen receptor degrader, as monotherapy and combined with abemaciclib, in recurrent/advanced ER-positive endometrioid endometrial cancer: Results from the phase 1a/1b EMBER study.
- Author
-
Yonemori K, Boni V, Min KG, Meniawy TM, Lombard J, Kaufman PA, Richardson DL, Bender L, Okera M, Matsumoto K, Giridhar KV, García-Sáenz JA, Prenen H, de Speville Uribe BD, Dizon DS, Garcia-Corbacho J, Van Nieuwenhuysen E, Li Y, Estrem ST, Nguyen B, Bacchion F, Ismail-Khan R, Jhaveri K, and Banda K
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Neoplasm Recurrence, Local drug therapy, Carcinoma, Endometrioid drug therapy, Carcinoma, Endometrioid pathology, Carcinoma, Endometrioid metabolism, Receptors, Estrogen metabolism, Aged, 80 and over, Aminopyridines administration & dosage, Aminopyridines adverse effects, Aminopyridines pharmacokinetics, Benzimidazoles administration & dosage, Benzimidazoles adverse effects, Benzimidazoles pharmacokinetics, Endometrial Neoplasms drug therapy, Endometrial Neoplasms pathology, Endometrial Neoplasms metabolism, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols administration & dosage
- Abstract
Objective: Imlunestrant is a next-generation oral selective estrogen receptor degrader designed to deliver continuous estrogen receptor (ER) target inhibition. EMBER is a phase 1a/b trial of imlunestrant, as monotherapy and combined with targeted therapy, in patients with ER+ advanced breast cancer or endometrioid endometrial cancer (EEC). This report focuses on patients with ER+ EEC., Methods: EMBER used an i3 + 3 dose-escalation design to determine the recommended phase 2 dose (RP2D) followed by dose-expansion cohorts (1:1 randomization): imlunestrant monotherapy and imlunestrant plus abemaciclib (150 mg twice daily). Eligible patients had measurable disease and progression or recurrence after platinum-containing chemotherapy. Prior fulvestrant or aromatase inhibitor was not allowed. Secondary endpoints included safety, pharmacokinetics and antitumor activity., Results: In total, 72 patients with a median of 2 prior anticancer therapies were treated. Among the 39 patients who received imlunestrant (400 mg [RP2D], n = 33; 800 mg, n = 6), the most common treatment-emergent adverse events (TEAEs) were grade 1-2 nausea (35.9 %), diarrhea (25.6 %), urinary tract infection (25.6 %), and abdominal pain (20.5 %). Overall response rate (ORR) was 10.3 %, clinical benefit rate (CBR) was 33.3 %, and median progression-free survival (mPFS) was 3.8 months (95 % CI, 1.8-6.7). Among the 33 patients who received imlunestrant (400 mg [RP2D], n = 29; 800 mg, n = 4) plus abemaciclib, the most common TEAEs were diarrhea (87.9 %), nausea (66.7 %), fatigue (48.5 %), and anemia (45.5 %). ORR was 18.2 %, CBR was 42.4 %, and mPFS was 6.8 months (95 % CI, 2.1-12)., Conclusion: Imlunestrant, as monotherapy and combined with abemaciclib, has a manageable safety profile with preliminary evidence of antitumor activity in patients with ER+ EEC., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Kan Yonemori: Institutional research support:MSD, Daiichi-Sankyo, Merck Biopharma, AstraZeneca, Taiho, Pfizer, Novartis, Takeda, Chugai, Ono, Sanofi, Seattle genetics, Eisai, Eli Lilly and company, Genmab, Boeringer Ingelheim, Kyowa Hakko Kirin, Nihon Kayaku, Haihe. Honoraria: Pfizer, Eisai, AstraZeneca, Eli Lilly and Company, Takeda, Chugai, Fuji Film Pharma, PDR pharma, MSD, Boeringer Ingelheim, Ono, Daiichi-Sankyo, Bayer, Jansen, Asteras, Bristol Myers Squibb, Novartis, Sanofi, Merk Biopharma. Advisory board: Eisai, Astra Zeneca, Sanofi, Genmab, Gliad, OncXerna, Takeda, Novartis, MSD, Henlius. Other: Principal investigator (to institution): MSD, Daiichi-Sankyo, Genmab, Seagen, AstraZeneca, Taiho, Merk Biopharma Pfizer, Novartis, Takeda, Chugai, Ono, Sanofi, Seattle genetics, Eisai, Eli Lilly and company, Genmab, Boeringer Ingelheim, Kyowa Hakko Kirin, Nihon Kayaku, Haihe. Valentina Boni: Institutional research support: Abbvie; ACEO; Adaptaimmune; Amcure; AMGEN; Amunix, AstraZeneca; Bycicle; BMS CytomX; GSK; Genentech/Roche; Genmab; Incyte; Ipsen; Janssen; Kura; Lilly; Loxo Therapeutics; Nektar; Macrogenics; Menarini; Merck; Merus; Nanobiotix; Novartis; Pfizer; PharmaMar; Principia; Puma; Ryvu; Ribbon; Sanofi; Taiho; Tesaro; BeiGene; Transgene; Takeda; Incyte; Innovio; MSD; PsiOxus; Seattle Genetics; Mersana; Daiichi; Nektar; Astellas; ORCA; Boston Therapeutics; Dynavax; DebioPharm; Boehringer Ingelheim; Regeneron; Rigontec; Millennium; Seagen; Synthon; Spectrum; Urogen; Zenith. Consulting fees: Puma Biotechnology; Ideaya Biosciences; Loxo Therapeutics, CytomX Therapeutics; Guidepoint; Oncoart, Lilly; Janssen; EMD Serono; IDMC Nanobiotix NANORAY-312/Novartis; Steering Committee CytomX Therapeutics. Honoraria (speaking): Eli Lilly and Company; MSD; SOLTI; TACTICS; Getthi; Gedefo. Attending meeting and/or travel: Bayer (ESMO GI). Stock/Ownership: 1TRIALSP. Tarek M. Meniawy: Research funding: Bristol Myers Squibb, AstraZeneca, Merck Serono, Roche, BeiGene, MSD, Regeneron, Incyte, GSK. Advisory board: Novartis, GSK, MSD, Sanofi, Eisai, BMS. Attending meeting and/or travel: Bristol Myers Squibb. Peter A. Kaufman: Consulting fees: Eli Lilly and Company. Honoraria: Eli Lilly and Company. Attending meeting and/or travel: Eli Lilly and Company. Debra L. Richardson: Advisory board: Mersana, AstraZeneca, GSK, Immunogen, Eisai, ProfoundBio, Daiichi-Sankyo. Koji Matsumoto: Research funding: Daichi-Sankyo, MSD, Eli Lilly and Company, Gilead Sciences, Eisai. Honoraria: MSD, Kyowa Kirin, Daichi-Sankyo, Eli Lilly and Company, Chugai. Karthik V. Giridhar: Research funding: Guardant Health; Advisory board: AstraZeneca, Eli Lilly and Company, Novartis, Neogenomics, TerSera Therapeutics, Puma Biotechnology. Attending meeting and/or travel: Eli Lilly and Company, Grail Inc. José Angel García-Sáenz: Grants or contracts: Stemline Menarini, Jazz Pharmaceutical. Consulting fees: Stemline Menarini, Eli Lilly and Company. Advisory Board: Daiichi Sankyo, AstraZeneca, Eli Lilly and Company, Novartis, Gilead. Speaker's bureau: Eli Lilly and Company, Novartis, Astrofarma. Research funding: AstraZeneca. Attending meeting and/or travel: Roche, Novartis, Daiichi-Sankyo. Don S. Dizon: Consulting fees: GSK, Clovis and Pharma, Astra Zeneca, Kronos Biotech. Stock: Doximity, Midi Health. Els Van Nieuwenhuysen: Research funding: Eli Lilly and Company. Yujia Li; Shawn T. Estrem; Bastien Nguyen; Francesca Bacchion; Roohi Ismail-Khan are employees and stock owners of Eli Lilly and Company. Komal Jhaveri: Institutional research funding:Novartis, Genentech, Debiopharm group, ADC therapeutics, Pfizer, Novita Pharmaceuticals, Clovis oncology, Eli Lilly and Company, Zymeworks, Immunomedics, Puma Biotechnology, VelosBio/Merck, AstraZeneca, Context Therapeutics, Scorpion Therapeutics, Blueprint Medicines. Consulting fees: Novartis, Pfizer, AstraZeneca, Jounce Therapeutics, Synthon, Intellisphere, Bristol Myers Squibb, Genetench, Abbvie, Eli Lilly and Company, BluePrint Medicines, Seagen, Daiichi-Sankyo, Biotheranostics, Sun Pharma Advanced Research Company, Taiho Oncology, Sanofi, Gilead Sciences, Scorpion Therapeutics. Attending meeting and/or travel: Taiho Pharmaceutical, Jounce therapeutics, Pfizer, AstraZeneca, Intellisphere, Eli Lilly and Company, Gilead Sciences, Genetench/Roche. Kalyan Banda: Institutional research funding: Eli Lilly and Company, MorphoSys, Regeneron, Kineta, Pfizer; Honoraria: American Society for Clinical Oncology; Advisory Board: Astra Zeneca, Immunogen; Attending meeting and/or travel: American Society for Clinical Oncology. The other authors have completed conflict of interest forms and do not have potential conflicts of interest related to subject matter discussed in this manuscript to declare., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
21. Lenvatinib plus pembrolizumab for patients with previously treated, advanced, triple-negative breast cancer: Results from the triple-negative breast cancer cohort of the phase 2 LEAP-005 Study.
- Author
-
Chung HC, Saada-Bouzid E, Longo F, Yanez E, Im SA, Castanon E, Desautels DN, Graham DM, Garcia-Corbacho J, Lopez J, Dutcus C, Okpara CE, Ghori R, Jin F, Groisberg R, and Korakis I
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Aged, 80 and over, Progression-Free Survival, Cohort Studies, Quinolines administration & dosage, Quinolines therapeutic use, Quinolines adverse effects, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology, Phenylurea Compounds administration & dosage, Phenylurea Compounds therapeutic use, Phenylurea Compounds adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
Background: Novel treatments are needed for patients with advanced, triple-negative breast cancer (TNBC) that progresses or recurs after first-line treatment with chemotherapy. The authors report results from the TNBC cohort of the multicohort, open-label, single-arm, phase 2 LEAP-005 study of lenvatinib plus pembrolizumab in patients with advanced solid tumors (ClinicalTrials.gov identifier NCT03797326)., Methods: Eligible patients had metastatic or unresectable TNBC with disease progression after one or two lines of therapy. Patients received lenvatinib (20 mg daily) plus pembrolizumab (200 mg every 3 weeks; up to 35 cycles). The primary end points were the objective response rate according to Response Evaluation Criteria in Solid Tumors, version 1.1, and safety (adverse events graded by the National Cancer Institute's Common Terminology Criteria for Adverse Events, version 4.0). Duration of response, progression-free survival, and overall survival were secondary end points., Results: Thirty-one patients were enrolled. The objective response rate by investigator assessment was 23% (95% confidence interval [CI], 10%-41%). Overall, the objective response rate by blinded independent central review (BICR) was 32% (95% CI, 17%-51%); and, in patients who had programmed cell death ligand 1 combined positive scores ≥10 (n = 8) and <10 (n = 22), the objective response rate was 50% (95% CI, 16%-84%) and 27% (95% CI, 11%-50%), respectively. The median duration of response by BICR was 12.1 months (range, from 3.0+ to 37.9+ months). The median progression-free survival by BICR was 5.1 months (95% CI, 1.9-11.8 months) and the median overall survival was 11.4 months (95% CI, 4.1-21.7 months). Treatment-related adverse events occurred in 94% of patients (grade 3, 52%; grade 4, 0%). One patient died due to a treatment-related adverse event of subarachnoid hemorrhage., Conclusions: The combination of lenvatinib plus pembrolizumab demonstrated antitumor activity with a manageable safety profile in patients with previously treated, advanced TNBC., (© 2024 Merck Sharp & Dohme LLC and The Author(s). Cancer published by Wiley Periodicals LLC on behalf of American Cancer Society.)
- Published
- 2024
- Full Text
- View/download PDF
22. Increasing Annual Cancer Incidence in Patients Age 20-49 Years: A Real-Data Study.
- Author
-
Ribelles N, Pascual J, Galvez-Carvajal L, Ruiz-Medina S, Garcia-Corbacho J, Benitez JC, Dominguez-Recio ME, Torres E, Oliva L, Zalabardo M, Rueda A, and Alba E
- Subjects
- Humans, Young Adult, Adult, Middle Aged, Incidence, Cross-Sectional Studies, Spain epidemiology, Lymphoma, Non-Hodgkin, Sarcoma
- Abstract
Purpose: Data from population-based studies have shown an increased incidence of certain types of neoplasms in patients younger than 50 years (early-onset cancer [EOC]); however, little information is derived from other real-world data sources. In a nonpopulation registry, we analyzed changes in the incidence of several neoplasms in successive generations., Methods: This cross-sectional study included all patients with a cancer diagnosis registered in one university hospital in Málaga, Spain, between 1998 and 2021, and 18 neoplasms were analyzed. For each neoplasm, the proportion of patients younger than 50 years and age 50 years and older (late-onset cancer [LOC]) of the total number of patients diagnosed each year was determined. In addition, the age limit was lowered to 45-40 years. Changes in these proportions between each year and the following year were assessed by calculating the annual percentage change (APC), and a final assessment of these changes was performed by determining the average APC (AAPC)., Results: Of the 24,596 patients, 5,466 (22.2%) had EOC, and 19,130 (77.8%) had LOC. The incidence of all tumors increased throughout the study period in both age groups. The AAPC increase was higher in patients with EOC than in those with LOC for the following neoplasms: head and neck (6.1% v 4.6%), colon (11.0% v 8.2%), testicular (16.3% v -13.1%), non-Hodgkin lymphoma (8.4% v 5.9%), rectum (16.1% v 6.8%), kidney (27.8% v 20.1%), and sarcoma (43.4% v 28.6%). This increase was confirmed in patients younger than 45 years and 40 years., Conclusion: Our results are consistent with the data published for most tumor sites analyzed. This global public health problem requires the utmost attention to decrease excess cancer in young patients.
- Published
- 2024
- Full Text
- View/download PDF
23. Praluzatamab Ravtansine, a CD166-Targeting Antibody-Drug Conjugate, in Patients with Advanced Solid Tumors: An Open-Label Phase I/II Trial.
- Author
-
Boni V, Fidler MJ, Arkenau HT, Spira A, Meric-Bernstam F, Uboha N, Sanborn RE, Sweis RF, LoRusso P, Nagasaka M, Garcia-Corbacho J, Jalal S, Harding JJ, Kim SK, Miedema IHC, Vugts DJ, Huisman MC, Zwezerijnen GJC, van Dongen GAMS, Menke van der Houven van Oordt CW, Wang S, Dang T, Zein IA, Vasiljeva O, Lyman SK, Paton V, Hannah A, and Liu JF
- Subjects
- Female, Humans, Tumor Microenvironment, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Immunoconjugates adverse effects, Maytansine therapeutic use, Neoplasms pathology
- Abstract
Purpose: Praluzatamab ravtansine (CX-2009) is a conditionally activated Probody drug conjugate (PDC) comprising an anti-CD166 mAb conjugated to DM4, with a protease-cleavable linker and a peptide mask that limits target engagement in normal tissue and circulation. The tumor microenvironment is enriched for proteases capable of cleaving the linker, thereby releasing the mask, allowing for localized binding of CX-2009 to CD166. CX-2009 was evaluated in a phase I/II clinical trial for patients with advanced solid tumors., Patients and Methods: Eligible patients had metastatic cancer receiving ≥2 prior treatments. CX-2009 was administered at escalating doses every 3 weeks (0.25-10 mg/kg) or every 2 weeks (4-6 mg/kg). Primary objective was to determine the safety profile and recommended phase II dose (RP2D)., Results: Of 99 patients enrolled, the most prevalent subtype was breast cancer (n = 45). Median number of prior therapies was 5 (range, 1-19). Dose-limiting toxicities were observed at 8 mg/kg every 3 weeks and 6 mg/kg every 2 weeks. On the basis of tolerability, the RP2D was 7 mg/kg every 3 weeks. Tumor regressions were observed at doses ≥4 mg/kg. In the hormone receptor-positive/HER2-nonamplified breast cancer subset (n = 22), 2 patients (9%) had confirmed partial responses, and 10 patients (45%) had stable disease. Imaging with zirconium-labeled CX-2009 confirmed uptake in tumor lesions and shielding of major organs. Activated, unmasked CX-2009 was measurable in 18 of 22 posttreatment biopsies., Conclusions: CD166 is a novel, ubiquitously expressed target. CX-2009 is the first conditionally activated antibody-drug conjugate to CD166 to demonstrate both translational and clinical activity in a variety of tumor types., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
24. Characteristics, origin, and potential for cancer diagnostics of ultrashort plasma cell-free DNA.
- Author
-
Hudecova I, Smith CG, Hänsel-Hertsch R, Chilamakuri CS, Morris JA, Vijayaraghavan A, Heider K, Chandrananda D, Cooper WN, Gale D, Garcia-Corbacho J, Pacey S, Baird RD, Rosenfeld N, and Mouliere F
- Subjects
- Biomarkers, Tumor genetics, DNA genetics, DNA, Single-Stranded, Humans, Sequence Analysis, DNA, Cell-Free Nucleic Acids genetics, Neoplasms diagnosis, Neoplasms genetics
- Abstract
Current evidence suggests that plasma cell-free DNA (cfDNA) is fragmented around a mode of 166 bp. Data supporting this view has been mainly acquired through the analysis of double-stranded cfDNA. The characteristics and diagnostic potential of single-stranded and damaged double-stranded cfDNA in healthy individuals and cancer patients remain unclear. Here, through a combination of high-affinity magnetic bead-based DNA extraction and single-stranded DNA sequencing library preparation (MB-ssDNA), we report the discovery of a large proportion of cfDNA fragments centered at ∼50 bp. We show that these "ultrashort" cfDNA fragments have a greater relative abundance in plasma of healthy individuals (median = 19.1% of all sequenced cfDNA fragments, n = 28) than in plasma of patients with cancer (median = 14.2%, n = 21, P < 0.0001). The ultrashort cfDNA fragments map to accessible chromatin regions of blood cells, particularly in promoter regions with the potential to adopt G-quadruplex (G4) DNA secondary structures. G4-positive promoter chromatin accessibility is significantly enriched in ultrashort plasma cfDNA fragments from healthy individuals relative to patients with cancers ( P < 0.0001), in whom G4-cfDNA enrichment is inversely associated with copy number aberration-inferred tumor fractions. Our findings redraw the landscape of cfDNA fragmentation by identifying and characterizing a novel population of ultrashort plasma cfDNA fragments. Sequencing of MB-ssDNA libraries could facilitate the characterization of gene regulatory regions and DNA secondary structures via liquid biopsy. Our data underline the diagnostic potential of ultrashort cfDNA through classification for cancer patients., (© 2022 Hudecova et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
25. Case Report: A Case Study Documenting the Activity of Atezolizumab in a PD-L1-Negative Triple-Negative Breast Cancer.
- Author
-
Brasó-Maristany F, Sansó M, Chic N, Martínez D, González-Farré B, Sanfeliu E, Ghiglione L, Carcelero E, Garcia-Corbacho J, Sánchez M, Soy D, Jares P, Peg V, Saura C, Muñoz M, Prat A, and Vivancos A
- Abstract
The immune checkpoint inhibitor atezolizumab is approved for PD-L1-positive triple-negative breast cancer (TNBC). However, no activity of atezolizumab in PD-L1-negative TNBC has been reported to date. Here, we present the case study of a woman with TNBC with low tumor infiltrating lymphocytes and PD-L1-negative disease, which achieved a significant response to atezolizumab monotherapy and durable response after the combination of atezolizumab and nab-paclitaxel. The comprehensive genomic analysis that we performed in her tumor and plasma samples revealed high tumor mutational burden (TMB), presence of the APOBEC genetic signatures, high expression of the tumor inflammation signature, and a HER2-enriched subtype by the PAM50 assay. Some of these biomarkers have been shown to independently predict response to immunotherapy in other tumors and may explain the durable response in our patient. Our work warrants further translational studies to identify biomarkers of response to immune checkpoint inhibitors in TNBC beyond PD-L1 expression and to better select patients that will benefit from immunotherapy., Competing Interests: Potential conflicts of interest are the following: AP reports consulting fees from Nanostring Technologies, Roche, Pfizer, Novartis, AstraZeneca, Foundation Medicine, Guardant Health, and Daiichi Sankyo outside the submitted work. AV reports consulting fees from Sysmex, Novartis, Merck, Bristol Meyers Squibb, Guardant Health, and Incyte; research funding from Bristol Meyers Squibb; and royalties from Ferrer outside the submitted work. VP has received fees as consultant, participated in advisory boards, or received travel grants from Sysmex, Roche, MSD, AstraZeneca, Bayer, and Exact Sciences outside the submitted work. CS has declared personal fees as consultant and advisory board or travel grants of AstraZeneca, Daiichi Sankyo, Eisai, Exact Sciences, Exeter Pharma, F. Hoffmann–La Roche Ltd, MediTech, Merck Sharp & Dohme, Novartis, Pfizer, Philips, Piere Fabre, Puma, Roche Farma, Sanofi-Aventis, SeaGen, and Zymeworks and institutional financial interests from AstraZeneca, Daiichi Sankyo, Eli Lilly and Company, Genentech, Immunomedics, Macrogenics, Merck, Sharp and Dhome España S.A., Novartis, Pfizer, Piqur Therapeutics, Puma, Roche, Synthon, and Zenith Pharma. CS and AP are Board Members of SOLTI Cooperative Group and are employed by Institut Oncològic Baselga (IOB), Quironsalud Group. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Brasó-Maristany, Sansó, Chic, Martínez, González-Farré, Sanfeliu, Ghiglione, Carcelero, Garcia-Corbacho, Sánchez, Soy, Jares, Peg, Saura, Muñoz, Prat and Vivancos.)
- Published
- 2021
- Full Text
- View/download PDF
26. Phase I, First-in-Human Study of the Probody Therapeutic CX-2029 in Adults with Advanced Solid Tumor Malignancies.
- Author
-
Johnson M, El-Khoueiry A, Hafez N, Lakhani N, Mamdani H, Rodon J, Sanborn RE, Garcia-Corbacho J, Boni V, Stroh M, Hannah AL, Wang S, Castro H, and Spira A
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Neoplasm Staging, Immunoconjugates therapeutic use, Neoplasms drug therapy, Neoplasms pathology
- Abstract
Purpose: PROCLAIM-CX-2029 is a phase I first-in-human study of CX-2029, a Probody-drug conjugate targeting CD71 (transferrin receptor 1) in adults with advanced solid tumors. Although the transferrin receptor is highly expressed across multiple tumor types, it has not been considered a target for antibody-drug conjugates (ADCs) due to its broad expression on normal cells. CX-2029 is a masked form of a proprietary anti-CD71 antibody conjugated to monomethyl auristatin E, designed to be unmasked in the tumor microenvironment by tumor-associated proteases, therefore limiting off-tumor toxicity and creating a therapeutic window for this previously undruggable target., Patients and Methods: This was a dose-escalation, multicenter trial to evaluate the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of CX-2029. The primary endpoint was to determine the maximum tolerated dose (MTD) and cycle 1 dose-limiting toxicity (DLT). CX-2029 was administered i.v. every 3 weeks., Results: Forty-five patients were enrolled in eight dose levels. No DLTs were reported in the dose escalation through 4 mg/kg. At 5 mg/kg, there were two DLTs (febrile neutropenia and pancytopenia). Following expansion of the 4 mg/kg dose to six patients, two additional DLTs were observed (infusion-related reaction and neutropenia/anemia). Both the 4 and 5 mg/kg doses were declared above the maximum tolerated dose. The recommended phase II dose is 3 mg/kg. The most common dose-dependent hematologic toxicities were anemia and neutropenia. Confirmed partial responses were observed in three patients, all with squamous histologies., Conclusions: The Probody therapeutic platform enables targeting CD71, a previously undruggable ADC target, at tolerable doses associated with clinical activity. See related commentary by Oberoi and Garralda, p. 4459 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
27. CX-072 (pacmilimab), a Probody ® PD-L1 inhibitor, in advanced or recurrent solid tumors (PROCLAIM-CX-072): an open-label dose-finding and first-in-human study.
- Author
-
Naing A, Thistlethwaite F, De Vries EGE, Eskens FALM, Uboha N, Ott PA, LoRusso P, Garcia-Corbacho J, Boni V, Bendell J, Autio KA, Randhawa M, Durm G, Gil-Martin M, Stroh M, Hannah AL, Arkenau HT, and Spira A
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Immune Checkpoint Inhibitors pharmacology, Male, Middle Aged, Young Adult, Immune Checkpoint Inhibitors therapeutic use, Immunotherapy methods, Neoplasms drug therapy
- Abstract
Background: Probody
® therapeutics are antibody prodrugs that are activated in the tumor microenvironment by tumor-associated proteases, thereby restricting the activity to the tumor microenvironment and minimizing 'off-tumor' toxicity. We report dose-escalation and single-agent expansion phase data from the first-in-human study of CX-072 (pacmilimab), a Probody checkpoint inhibitor directed against programmed death-ligand 1 (PD-L1)., Methods: In the dose-escalation phase of this multicenter, open-label study (NCT03013491), adults with advanced solid tumors (naive to programmed-death-1/PD-L1 or cytotoxic T-lymphocyte-associated antigen 4 inhibitors) were enrolled into one of seven dose-escalation cohorts, with pacmilimab administered intravenously every 14 days. The primary endpoints were safety and determination of the maximum tolerated dose (MTD). In the expansion phase, patients with one of six prespecified malignancies (triple-negative breast cancer [TNBC]; anal squamous cell carcinoma [aSCC]; cutaneous SCC [cSCC]; undifferentiated pleomorphic sarcoma [UPS]; small bowel adenocarcinoma [SBA]; and thymic epithelial tumor [TET]); or high tumor mutational burden (hTMB) tumors were enrolled. The primary endpoint was objective response (Response Evaluation Criteria In Solid Tumors v.1.1)., Results: An MTD was not reached with doses up to 30 mg/kg. A recommended phase 2 dose (RP2D) of 10 mg/kg was chosen based on pharmacokinetic and pharmacodynamic findings in the expansion phase. Ninety-eight patients enrolled in the expansion phase: TNBC (n=14), aSCC (n=14), cSCC (n=14), UPS (n=20), SBA (n=14), TET (n=8), and hTMB tumors (n=14). Of 114 patients receiving pacmilimab at the RP2D, grade ≥3 treatment-related adverse events (TRAEs) were reported in 10 patients (9%), serious TRAEs in six patients (5%), and treatment discontinuation due to TRAEs in two patients (2%). Grade ≥3 immune-related AEs occurred in two patients (rash, myocarditis). High PD-L1 expression (ie, >50% Tumor Proportion Score) was observed in 22/144 (19%) patients. Confirmed objective responses were observed in patients with cSCC (n=5, including one complete response), hTMB (n=4, including one complete response), aSCC (n=2), TNBC (n=1), UPS (n=1), and anaplastic thyroid cancer (n=1)., Conclusions: Pacmilimab can be administered safely at the RP2D of 10 mg/kg every 14 days. At this dose, pacmilimab had a low rate of immune-mediated toxicity and showed signs of antitumor activity in patients not selected for high PD-L1 expression., Trial Registration Number: NCT03013491., Competing Interests: Competing interests: AN: Research funding from Amplimmune, Arcus Biosciences, ARMO BioSciences, Atterocor, BMS, Calithera Biosciences, CytomX Therapeutics, Eli Lilly, EMD, HealiosOnc, ImmuneOncia, Incyte, Karyopharm Therapeutics, Kymab, MedImmune, Merck, NCI, NeoimmuneTech, Neon Therapeutics, Novartis, Nutrition, Pfizer, PsiOxus, Regeneron, Serono, Surface Oncology, and TopAlliance Biosciences. Advisory boards for CytomX Therapeutics, Genome & Company, Kymab, Novartis, OncoSec KEYNOTE-695, and STCube Pharmaceuticals. Travel expenses from ARMO BioSciences. AN’s spouse has received research funding from Baxalta, Chao physician-scientist, Immune Deficiency Foundation, and Jeffery Modell Foundation; and has served on advisory boards for Behring, CSL, Horizon, Pharming, and Takeda. FT: Research funding and conference registration from Novartis. Consultancy/advisory role for Achilles Therapeutics, Bayer, BMS, Enara Bio, GSK, T-Knife, and Zelluna. EGEDV: Institutional financial support for clinical trials or contracted research from Amgen, AstraZeneca, Bayer, Chugai Pharma, G1 Therapeutics, Genentech, Nordic Nanovector, Radius Health, Regeneron, Roche, Servier, and Synthon. Institutional financial support for advisory boards from Daiichi Sankyo, Merck, NSABP, Pfizer, and Sanofi; all outside the submitted work. FALME: Consultancy/advisory role for Servier, Novartis, Eisai, and Ipsen. NU: Consultant for AstraZeneca, Eli Lilly, Ipsen, QED, and Taiho. Research support from EMD, Ipsen, Serono, and Taiho. Holds stock in Exact Sciences and Natera. PAO: Research funding from and advisory role for Amgen, Armo BioSciences, Array, AstraZeneca/MedImmune, Bristol-Meyers Squibb, Celldex, CytomX, Merck, Neon Therapeutics, Novartis, Pfizer, and Roche/Genentech. PL: Consultant/advisory board for AbbVie, ABL Bio, Agenus, Agios, Astellas, AstraZeneca, Black Diamond, Cybrexa, CytomX, EMD Serono, GenMab, Genentech, Glaxo-Smith Kline, ImmunoMet, IQVIA, Kineta Inc., Kyowa Kirin Pharmaceutical Development, MacroGenics, Molecular Templates, Pfizer, QED Therapeutics, Salarius, Shattuck, Silverback, SK Life Science, SOTIO, STCube Pharmaceuticals, Takeda, TRIGR, and Zentalis Pharmaceuticals. Data safety monitoring committee for Five Prime, Halozyme, and Tyme. Participant in imCORE Alliance (Roche/Genentech). JG-C: Speaker’s bureau for Bayer; fees to support registration and attending scientific meetings from BMS and Novartis. VB: Consulting/advisory role for CytomX Therapeutics, Guidepoint, Ideaya Biosciences, Loxo Therapeutics, Oncoart, and Puma Biotechnology. Institutional financial support for clinical trials from Abbvie, ACEO, Adaptaimmune, Amcure, AMGEN, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Boston Therapeutics, Cytomx Therapeutics, Daiichi, DebioPharm, Dynavax, GSK, Genentech/Roche, H3, Incyte, Innovio, Janssen, Kura, Lilly, Loxo, Macrogenics, Menarini, Merck, Mersana, Merus, Millenium, MSD, Nanobiotix, Nektar, Novartis, ORCA, Pfizer, PharmaMar, Principia, PsiOxus, PUMA, Regeneron, Rigontec, Sanofi, Seattle Genetics, Spectrum, Synthon, Taiho, Tesaro, Transgene, Takeda, and Zenith. JB: Institutional financial support for clinical trials or contracted research from AbbVie, Acerta Pharma, ADC, Agios, Amgen, Apexigen, Arch Oncology, Arcus Bio, ARMO, Array, Arrys, AstraZeneca, AtlasMedx, Bayer, Beigene, Bellicum, BI, Bicycle Therapeutics, Blueprint, BMS, Boston Biomedical, CALGB, Calithera, Celgene, Celldex, Cyteir Therapeutics, Cytomx, Daiichi Sankyo, Effector, Eisai, EMD Serono, Evelo, Five Prime, FORMA, Forty Seven, Foundation Bio, Genentech/Roche, Gilead, Gossamer Bio, GSK, Harpoon, Hutchinson MediPharma, IGM Biosciences, Imclone, Incyte, Innate, Innate Pharma, Ipsen, Jacobio, Koltan, LEAP, Lilly, Mabspace, Macrogenics, Marshall Edwards, MedImmune, Merck, Merrimack, Mersana, Merus, Millennium, Morphotex, Nektar, NeoImmune Tech, NGM Biopharma, Novartis, Novocare, NuMab, Oncogenex, OncoMed, Ongologie, Onyx, Pfizer, Pieris, Prelude Oncology, PureTech Health, Regeneron, Relay Therapeutics, REPARE Therapeutics, Revolution Medicines, Rgenix, Sanofi, Scholar Rock, Seattle Genetics, Shattuck Labs, Sierra, Stemcentrx, SynDevRex, Synthorx, Taiho, Takeda, Tarveda, TempestTx, TG Therapeutics, Tracon, Treadwell Therapeutics, Tyrogenex, Unum Therapeutics, Vyriad, and Zymeworks. Institutional financial support for advisory boards/consulting from Array, Agios, Amgen, Apexigen, Arch Oncology, ARMO, AstraZeneca, Bayer, Beigene, BI, Bicycle Therapeutics, BMS, Celgene, Continuum Clinical, Cyteir, Daiichi Sankyo, Evelo, Five Prime, FORMA, Fusion Therapeutics, Genentech/Roche, Gilead, GSK, Incyte, Innate, Ipsen, Janssen, LEAP, Lilly, Macrogenics, MedImmune, Merck, Merrimack, Moderna Therapeutics, Molecular Partners, Novartis, Oncogenex, OncoMed, Pfizer, Phoenix Bio, Piper Biotech, Prelude Therapeutics, Relay Therapeutics, Samsung Bioepios, Sanofi, Seattle Genetics, Taiho, Tanabe Research Laboratories, TD2 (Translational Drug Development), TG Therapeutics, Tizona, Tolero, and Torque. Food, beverages and/or travel expenses from ARMO, BI, BMS, Celgene, FORMA, Genentech/Roche, Gilead, Ipsen, Lilly, MedImmune, Merck, Novartis, Oncogenex, OncoMed, and Taiho. KAA: Advisory board for CytomX (unpaid). Institutional financial support for clinical trials or contracted research from Amgen, AstraZeneca, CytomX, GSK, Merck, Pfizer, and Tizona. MR: Honoraria from Bayer. GD: Research funding from AstraZeneca, Bristol Myers Squibb, and Merck. Honoraria from AstraZeneca. Advisory board for Curio Science. MG-M: Speaker’s bureau for Astra Zeneca, Pharmamar, and Roche. Financial support for registration and attendance at scientific meetings from MDS, Pharmamar, and Roche. MS is a former employee and ALH is a current employee of CytomX Therapeutics, Inc. Both are stockowners in CytomX Therapeutics Inc. H-TA: Advisor for Bayer, Beigene, Bicycle, Engitix, Guardant, iOnctura, Roche, and Servier. Employed by HCA Healthcare UK and Sarah Cannon Research Institute. AS: Consultant for Amgen, AstraZeneca, BMS, Merck, Mirati, and Novartis. Institutional financial support for clinical trials or contracted research from CytomX., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
28. CX-072 (pacmilimab), a Probody PD-L1 inhibitor, in combination with ipilimumab in patients with advanced solid tumors (PROCLAIM-CX-072): a first-in-human, dose-finding study.
- Author
-
Sanborn RE, Hamid O, de Vries EG, Ott PA, Garcia-Corbacho J, Boni V, Bendell J, Autio KA, Cho DC, Plummer R, Stroh M, Lu L, and Thistlethwaite F
- Subjects
- Adult, Aged, Antibodies, Monoclonal administration & dosage, B7-H1 Antigen immunology, Dose-Response Relationship, Drug, Female, Humans, Immune Checkpoint Inhibitors administration & dosage, Immunotherapy, Ipilimumab administration & dosage, Male, Middle Aged, Neoplasms immunology, Prodrugs administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, B7-H1 Antigen antagonists & inhibitors, Neoplasms drug therapy
- Abstract
Background: Probody® therapeutics are antibody prodrugs designed to be activated by tumor-associated proteases. This conditional activation restricts antibody binding to the tumor microenvironment, thereby minimizing 'off-tumor' toxicity. Here, we report the phase 1 data from the first-in-human study of CX-072 (pacmilimab), a Probody immune checkpoint inhibitor directed against programmed death-ligand 1 (PD-L1), in combination with the anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) antibody ipilimumab., Methods: Adults (n=27) with advanced solid tumors (naive to PD-L1/programmed cell death protein 1 or CTLA-4 inhibitors) were enrolled in the phase 1 combination therapy dose-escalation portion of this multicenter, open-label, phase 1/2 study (NCT03013491). Dose-escalation pacmilimab/ipilimumab followed a standard 3+3 design and continued until the maximum tolerated dose (MTD) was determined. Pacmilimab+ipilimumab was administered intravenously every 3 weeks for four cycles, followed by pacmilimab administered every 2 weeks as monotherapy. The primary objective was identification of dose-limiting toxicities and determination of the MTD. Other endpoints included the rate of objective response (Response Evaluation Criteria In Solid Tumors v.1.1)., Results: Twenty-seven patients were enrolled in pacmilimab (mg/kg)+ipilimumab (mg/kg) dose-escalation cohorts: 0.3+3 (n=6); 1+3 (n=3); 3+3 (n=3); 10+3 (n=8); 10+6 (n=6); and 10+10 (n=1). Dose-limiting toxicities occurred in three patients, one at the 0.3+3 dose level (grade 3 dyspnea/pneumonitis) and two at the 10+6 dose level (grade 3 colitis, grade 3 increased aspartate aminotransferase). The MTD and recommended phase 2 dose was pacmilimab 10 mg/kg+ipilimumab 3 mg/kg administered every 3 weeks. Pacmilimab-related grade 3-4 adverse events (AEs) and grade 3-4 immune-related AEs were reported in nine (33%) and six (22%) patients, respectively. Three patients (11%) discontinued treatment because of AEs. The overall response rate was 19% (95% CI 6.3 to 38.1), with one complete (anal squamous cell carcinoma) and four partial responses (cancer of unknown primary, leiomyosarcoma, mesothelioma, testicular cancer). Responses lasted for >12 months in four patients., Conclusions: The MTD and recommended phase 2 dose of pacmilimab (10 mg/kg)+ipilimumab (3 mg/kg) every 3 weeks is active and has a favorable tolerability profile., Competing Interests: Competing interests: RES: Honoraria from Amgen and AstraZeneca. Advisory boards for AstraZeneca, Blueprint Medicines, Celldex, Daiichi Sankyo, EMD Serono, Genentech/Roche, Janssen, and Seagen. Research support from AstraZeneca and Merck. OH: Consulting/advisory boards for Aduro, Akeso, Amgen, Beigene, Bioatla, BMS, Roche Genentech, GSK, lmmunocore, ldera, lncyte, Janssen, Merck, Nextcure, Novartis, Pfizer, Sanofi/Regeneron, Seattle Genetics, Tempus, and Zelluna. Speakers’ bureau for BMS, Novartis, Pfizer, and Sanofi/Regeneron. Institutional financial support for contracted research from Arcus, Aduro, Akeso, Amgen, Bioatla, BMS, CytomX, Exelixis, Roche Genentech, GSK, lmmunocore, ldera, lncyte, lovance, Merck, Moderna, Merck-Serano, NextCure, Novartis, Pfizer, Sanofi/Regeneron, Seattle Genetics, Torque, and Zelluna. EGEdV: Institutional financial support for clinical trials or contracted research from Amgen, AstraZeneca, Bayer, Chugai Pharma, G1 Therapeutics Genentech, Nordic Nanovector, Radius Health, Regeneron, Roche, Servier, and Synthon. Institutional financial support for advisory boards from Daiichi Sankyo, Merck, NSABP, Pfizer, and Sanofi; all outside the submitted work. PAO: Research funding from and has advisory role for Neon Therapeutics, Bristol-Meyers Squibb, Merck, CytomX, Pfizer, Novartis, Celldex, Amgen, Array, AstraZeneca/MedImmune, Armo BioSciences and Roche/Genentech. JG-C: Speaker’s bureau for Bayer; Fees to support registration and attending scientific meetings from BMS, Novartis. VB: Consulting/advisory role for CytomX Therapeutics, Guidepoint, Ideaya Biosciences; Loxo Therapeutics, Oncoart, and Puma Biotechnology. Institutional financial support for clinical trials from Abbvie, ACEO, Adaptaimmune, Amcure, Amgen, Astellas, AstraZeneca, BeiGene, BMS, Boehringer Ingelheim, Boston Therapeutics, CytomX Therapeutics, Daiichi, DebioPharm, Dynavax, GSK, Genentech/Roche, H3, Incyte, Innovio, Janssen, Kura, Lilly, Loxo, Mersana, Nektar, Macrogenics, Menarini, Merck, Merus, Millenium, MSD, Nanobiotix, Novartis, ORCA, Pfizer, PharmaMar, Principia, PsiOxus, PUMA, Regeneron, Rigontec, Sanofi, Seattle Genetics, Spectrum, Synthon, Taiho, Tesaro, Transgene, Takeda, and Zenith. JB: Institutional financial support for clinical trials or contracted research from AbbVie, Acerta Pharma, ADC, Agios, Amgen, Apexigen, Arch Oncology, Arcus Bio, ARMO, Array, Arrys, AstraZeneca, AtlasMedx, Bayer, Beigene, Bellicum, BI, Bicycle Therapeutics, Blueprint, BMS, Boston Biomedical, CALGB, Calithera, Celgene, Celldex, Cyteir Therapeutics, Cytomx, Daiichi Sankyo, Effector, Eisai, EMD Serono, Evelo, Five Prime, FORMA, Forty Seven, Foundation Bio, Genentech / Roche, Gilead, Gossamer Bio, GSK, Harpoon, Hutchinson MediPharma, IGM Biosciences, Imclone, Incyte, Innate, Innate Pharma, Ipsen, Jacobio, Koltan, LEAP, Lilly, Mabspace, Macrogenics, Marshall Edwards, MedImmune, Merck, Merrimack, Mersana, Merus, Millennium, Morphotex, Nektar, NeoImmune Tech, NGM Biopharma, Novartis, Novocare, NuMab, Oncogenex, OncoMed, Ongologie, Onyx, Pfizer, Pieris, Prelude Oncology, PureTech Health, Regeneron, Relay Therapeutics, REPARE Therapeutics, Revolution Medicines, Rgenix, Sanofi, Scholar Rock, Seattle Genetics, Shattuck Labs, Sierra, Stemcentrx, SynDevRex, Synthorx, Taiho, Takeda, Tarveda, TempestTx, TG Therapeutics, Tracon, Treadwell Therapeutics, Tyrogenex, Unum Therapeutics, Vyriad, and Zymeworks. Institutional financial support for advisory boards/consulting from Array, Agios, Amgen, Apexigen, Arch Oncology, ARMO, AstraZeneca, Bayer, Beigene, BI, Bicycle Therapeutics, BMS, Celgene, Continuum Clinical, Cyteir, Daiichi Sankyo, Evelo, Five Prime, FORMA, Fusion Therapeutics, Genentech / Roche, Gilead, GSK, Incyte, Innate, Ipsen, Janssen, LEAP, Lilly, Macrogenics, MedImmune, Merck, Merrimack, Moderna Therapeutics, Molecular Partners, Novartis, Oncogenex, OncoMed, Pfizer, Phoenix Bio, Piper Biotech, Prelude Therapeutics, Relay Therapeutics, Samsung Bioepios, Sanofi, Seattle Genetics, Taiho, Tanabe Research Laboratories, TD2 (Translational Drug Development), TG Therapeutics, Tizona, Tolero, and Torque. Food, beverages and/or travel expenses from ARMO, BI, BMS, Celgene, FORMA, Genentech / Roche, Gilead, Ipsen, Lilly, MedImmune, Merck, Novartis, Oncogenex, OncoMed, and Taiho. KAA: Advisory board for CytomX (unpaid). Institutional financial support for clinical trials or contracted research from Amgen, AstraZeneca, CytomX, GSK, Merck, Pfizer, and Tizona. DCC: Consultant for Nektar Therapeutics, GSK, Torque, PureTech, and HUYA. RP: Honoria for attending advisory boards from Pierre Faber, Bayer, Octimet, Clovis Oncology, Novartis, Karus Therapeutics, Biosceptre, BMS, Cybrexa, Ellipses, CV6 Therapeutics, Astex Therapeutics and Sanofi Aventis. Fees for delivery of educational talks or chairing educational meetings by AstraZeneca, Novartis, Bayer, Tesaro and BMS. Funds to support attendance at conferences from BMS and MSD. MS is a former employee and Lawrence Lu is a current employee of CytomX Therapeutics and both own stock in CytomX Therapeutics. FT: Research funding and conference registration from Novartis. Consultancy/advisory role for GSK, Achilles Therapeutics, BMS, Zelluna, Enara Bio, Bayer, T-Knife., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
29. Assessment of the Feasibility and Safety of Durvalumab for Treatment of Solid Tumors in Patients With HIV-1 Infection: The Phase 2 DURVAST Study.
- Author
-
Gonzalez-Cao M, Morán T, Dalmau J, Garcia-Corbacho J, Bracht JWP, Bernabe R, Juan O, de Castro J, Blanco R, Drozdowskyj A, Argilaguet J, Meyerhans A, Blanco J, Prado JG, Carrillo J, Clotet B, Massuti B, Provencio M, Molina-Vila MA, Mayo de Las Casa C, Garzon M, Cao P, Huang CY, Martinez-Picado J, and Rosell R
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Antineoplastic Agents, Immunological adverse effects, Female, HIV-1, Humans, Male, Middle Aged, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents, Immunological therapeutic use, HIV Infections drug therapy, Neoplasms drug therapy
- Abstract
Importance: Therapies targeting the programmed cell death 1 (PD-1) receptor or its ligand (PD-L1), such as the humanized monoclonal antibody durvalumab, have shown durable clinical responses in several tumor types. However, concerns about the safety and feasibility of PD-1/PD-L1 blockade in HIV-1-infected individuals have led to the exclusion of these patients from clinical trials on cancer immunotherapies., Objective: To evaluate the feasibility and safety of durvalumab treatment in patients with advanced cancer and virologically controlled HIV-1 infection., Design, Setting, and Participants: The DURVAST study was a nonrandomized, open-label, phase 2 clinical trial in patients with any solid tumor type in which anti-PD-1 or anti-PD-L1 antibodies have approved indications or for which there are data of antitumoral activity with no other available curative therapy. All patients had basal undetectable plasma viremia while undergoing combination antiretroviral therapy., Interventions: Treatment consisted of intravenous infusion of durvalumab (1500 mg every 4 weeks) until disease progression or unacceptable toxic effects., Main Outcomes and Measures: Adverse events were graded with the use of the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.03. Tumor response was evaluated using the Response Evaluation Criteria in Solid Tumors version 1.1., Results: A total of 20 HIV-1-infected patients with advanced cancer were enrolled; 16 (80%) were male, the median (range) age was 54 (30-73) years, and 12 (60%) had progressed with previous cancer treatment lines. A median (range) of 4 (1-16) cycles of durvalumab were administered. Drug-related adverse events were observed in 50% of patients, and all were grade 1 and 2 (mainly diarrhea, asthenia, and arthromyalgia). Four of 16 response-evaluable patients (25%) had a partial response. Five patients (31%) had stable disease, including 4 with durable stable disease (disease control rate of 50%). CD4+ and CD8+ T-cell counts and plasma HIV-1 viremia remained stable throughout the study., Conclusions and Relevance: Durvalumab treatment was feasible and safe in HIV-1-infected patients with cancer receiving combination antiretroviral therapy. HIV-1-infected patients on suppressive antiretroviral therapy with advanced cancer should have access to cancer immunotherapy treatments., Trial Registration: ClinicalTrials.gov Identifier: NCT03094286.
- Published
- 2020
- Full Text
- View/download PDF
30. A phase I/II study of epertinib plus trastuzumab with or without chemotherapy in patients with HER2-positive metastatic breast cancer.
- Author
-
Macpherson IR, Spiliopoulou P, Rafii S, Saggese M, Baird RD, Garcia-Corbacho J, Italiano A, Bonneterre J, Campone M, Cresti N, Posner J, Takeda Y, Arimura A, and Spicer J
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Brain Neoplasms drug therapy, Brain Neoplasms secondary, Breast Neoplasms pathology, Capecitabine administration & dosage, Capecitabine adverse effects, Capecitabine therapeutic use, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Humans, Middle Aged, Quinazolines administration & dosage, Quinazolines adverse effects, Receptor, ErbB-2 antagonists & inhibitors, Trastuzumab administration & dosage, Trastuzumab adverse effects, Treatment Outcome, Vinorelbine administration & dosage, Vinorelbine adverse effects, Vinorelbine therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Quinazolines therapeutic use, Receptor, ErbB-2 metabolism, Trastuzumab therapeutic use
- Abstract
Background: Epertinib (S-222611) is a potent reversible inhibitor of HER2, EGFR and HER4. This trial evaluated the safety, tolerability, pharmacokinetics and antitumour activity of daily oral epertinib combined with trastuzumab (arm A), with trastuzumab plus vinorelbine (arm B) or with trastuzumab plus capecitabine (arm C), in patients with HER2-positive metastatic breast cancer (MBC)., Methods: Eligible patients, with or without brain metastases, had received prior HER2-directed therapy. A dose-escalation phase determined the tolerability of each combination and established a dose for further study. Further, patients were recruited to expansion cohorts in each of the 3 arms to further explore efficacy and safety., Results: The recommended doses of epertinib were 600 mg, 200 mg and 400 mg in arms A, B and C, respectively. The most frequent grade 3/4 adverse event (AE) was diarrhoea in all arms, which was manageable with medical intervention and dose modification. The objective response rate (complete response [CR] plus partial response [PR]) in heavily pre-treated HER2-positive MBC patients at the recommended doses of epertinib combined with trastuzumab was 67% (N = 9), with trastuzumab plus vinorelbine was 0% (N = 5) and with trastuzumab plus capecitabine was 56% (N = 9). Notably, 4 of 6 patients previously treated with T-DM1 responded in the arm A expansion cohort (epertinib plus trastuzumab). In the arm C expansion cohort (epertinib plus trastuzumab plus capecitabine), 4 of 7 patients responded despite previous exposure to capecitabine. Measurable regression of brain metastases was observed in patients with CNS target lesions treated in both arms A and C., Conclusion: We observed safety, tolerability and encouraging antitumour activity of epertinib combined with trastuzumab, or with trastuzumab plus capecitabine. This supports further evaluation of these combinations in patients with pre-treated HER2-positive MBC, with or without brain metastases., Trial Registration: EudraCT Number: 2013-003894-87; registered 09-September-2013.
- Published
- 2019
- Full Text
- View/download PDF
31. POSEIDON Trial Phase 1b Results: Safety, Efficacy and Circulating Tumor DNA Response of the Beta Isoform-Sparing PI3K Inhibitor Taselisib (GDC-0032) Combined with Tamoxifen in Hormone Receptor Positive Metastatic Breast Cancer Patients.
- Author
-
Baird RD, van Rossum AGJ, Oliveira M, Beelen K, Gao M, Schrier M, Mandjes IAM, Garcia-Corbacho J, Vallier AL, Dougall G, van Werkhoven E, Linossi C, Kumar S, van Tinteren H, Callari M, Beddowes E, Perez-Garcia JM, Rosing H, Platte E, Nederlof P, Schot M, de Vries Schultink A, Bernards R, Saura C, Gallagher W, Cortès J, Caldas C, and Linn SC
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms diagnosis, Breast Neoplasms mortality, Female, Humans, Imidazoles administration & dosage, Imidazoles pharmacokinetics, Middle Aged, Mutation, Neoplasm Metastasis, Neoplasm Staging, Oxazepines administration & dosage, Oxazepines pharmacokinetics, Receptors, Estrogen genetics, Receptors, Progesterone genetics, Retreatment, Tamoxifen administration & dosage, Tamoxifen pharmacokinetics, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Circulating Tumor DNA
- Abstract
Purpose: The strategy of combining endocrine therapy with PI3K-mTOR inhibition has shown promise in estrogen receptor (ER)-positive breast cancer, but new agents and combinations with a better therapeutic index are urgently needed. Taselisib is a potent, selective, beta-isoform-sparing PI3 kinase inhibitor., Patients and Methods: 30 patients with ER-positive, metastatic breast cancer who had failed prior endocrine therapy were treated with escalating doses of taselisib (2 or 4 mg in an intermittent or continuous schedule) combined with tamoxifen 20 mg once daily in this phase 1b study using a "rolling six" design., Results: Taselisib combined with tamoxifen was generally well tolerated, with treatment-emergent adverse events as expected for this class of drugs, including diarrhea (13 patients, 43%), mucositis (10 patients, 33%), and hyperglycemia (8 patients, 27%). No dose-limiting toxicities were observed. Objective responses were seen in 6 of 25 patients with RECIST-measurable disease (ORR 24%). Median time to disease progression was 3.7 months. Twelve of 30 patients (40%) had disease control for 6 months or more. Circulating tumor (ct)DNA studies using next-generation tagged amplicon sequencing identified early indications of treatment response and mechanistically relevant correlates of clinical drug resistance (e.g., mutations in KRAS, ERBB2 ) in some patients., Conclusions: Taselisib can be safely combined with tamoxifen at the recommended phase 2 dose of 4 mg given once daily on a continuous schedule. Preliminary evidence of antitumor activity was seen in both PIK3CA mutant and wild-type cancers. The randomized phase 2 part of POSEIDON (testing tamoxifen plus taselisib or placebo) is currently recruiting., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
32. Enhanced detection of circulating tumor DNA by fragment size analysis.
- Author
-
Mouliere F, Chandrananda D, Piskorz AM, Moore EK, Morris J, Ahlborn LB, Mair R, Goranova T, Marass F, Heider K, Wan JCM, Supernat A, Hudecova I, Gounaris I, Ros S, Jimenez-Linan M, Garcia-Corbacho J, Patel K, Østrup O, Murphy S, Eldridge MD, Gale D, Stewart GD, Burge J, Cooper WN, van der Heijden MS, Massie CE, Watts C, Corrie P, Pacey S, Brindle KM, Baird RD, Mau-Sørensen M, Parkinson CA, Smith CG, Brenton JD, and Rosenfeld N
- Subjects
- Animals, Circulating Tumor DNA blood, DNA Copy Number Variations genetics, Genome, Human, Humans, Machine Learning, Mice, Mutation genetics, Whole Genome Sequencing, Circulating Tumor DNA analysis, Circulating Tumor DNA chemistry
- Abstract
Existing methods to improve detection of circulating tumor DNA (ctDNA) have focused on genomic alterations but have rarely considered the biological properties of plasma cell-free DNA (cfDNA). We hypothesized that differences in fragment lengths of circulating DNA could be exploited to enhance sensitivity for detecting the presence of ctDNA and for noninvasive genomic analysis of cancer. We surveyed ctDNA fragment sizes in 344 plasma samples from 200 patients with cancer using low-pass whole-genome sequencing (0.4×). To establish the size distribution of mutant ctDNA, tumor-guided personalized deep sequencing was performed in 19 patients. We detected enrichment of ctDNA in fragment sizes between 90 and 150 bp and developed methods for in vitro and in silico size selection of these fragments. Selecting fragments between 90 and 150 bp improved detection of tumor DNA, with more than twofold median enrichment in >95% of cases and more than fourfold enrichment in >10% of cases. Analysis of size-selected cfDNA identified clinically actionable mutations and copy number alterations that were otherwise not detected. Identification of plasma samples from patients with advanced cancer was improved by predictive models integrating fragment length and copy number analysis of cfDNA, with area under the curve (AUC) >0.99 compared to AUC <0.80 without fragmentation features. Increased identification of cfDNA from patients with glioma, renal, and pancreatic cancer was achieved with AUC > 0.91 compared to AUC < 0.5 without fragmentation features. Fragment size analysis and selective sequencing of specific fragment sizes can boost ctDNA detection and could complement or provide an alternative to deeper sequencing of cfDNA., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
33. Association between PD1 mRNA and response to anti-PD1 monotherapy across multiple cancer types.
- Author
-
Paré L, Pascual T, Seguí E, Teixidó C, Gonzalez-Cao M, Galván P, Rodríguez A, González B, Cuatrecasas M, Pineda E, Torné A, Crespo G, Martin-Algarra S, Pérez-Ruiz E, Reig Ò, Viladot M, Font C, Adamo B, Vidal M, Gaba L, Muñoz M, Victoria I, Ruiz G, Viñolas N, Mellado B, Maurel J, Garcia-Corbacho J, Molina-Vila MÁ, Juan M, Llovet JM, Reguart N, Arance A, and Prat A
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, CD8-Positive T-Lymphocytes immunology, Cohort Studies, Female, Follow-Up Studies, Humans, Lymphocytes, Tumor-Infiltrating immunology, Male, Middle Aged, Neoplasms drug therapy, Neoplasms immunology, Neoplasms pathology, Prognosis, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor genetics, RNA, Messenger genetics, Survival Rate, Antineoplastic Agents, Immunological therapeutic use, CD8-Positive T-Lymphocytes drug effects, Gene Expression Regulation, Neoplastic drug effects, Lymphocytes, Tumor-Infiltrating drug effects, Neoplasms metabolism, Programmed Cell Death 1 Receptor metabolism, RNA, Messenger metabolism
- Abstract
Background: We hypothesized that the abundance of PD1 mRNA in tumor samples might explain the differences in overall response rates (ORR) observed following anti-PD1 monotherapy across cancer types., Patients and Methods: RNASeqv2 data from 10 078 tumor samples representing 34 different cancer types was analyzed from TCGA. Eighteen immune-related gene signatures and 547 immune-related genes, including PD1, were explored. Correlations between each gene/signature and ORRs reported in the literature following anti-PD1 monotherapy were calculated. To translate the in silico findings to the clinical setting, we analyzed the expression of PD1 mRNA using the nCounter platform in 773 formalin-fixed paraffin embedded (FFPE) tumor samples across 17 cancer types. To test the direct relationship between PD1 mRNA, PDL1 immunohistochemistry (IHC), stromal tumor-infiltrating lymphocytes (sTILs) and ORR, we evaluated an independent FFPE-based dataset of 117 patients with advanced disease treated with anti-PD1 monotherapy., Results: In pan-cancer TCGA, PD1 mRNA expression was found strongly correlated (r > 0.80) with CD8 T-cell genes and signatures and the proportion of PD1 mRNA-high tumors (80th percentile) within a given cancer type was variable (0%-84%). Strikingly, the PD1-high proportions across cancer types were found strongly correlated (r = 0.91) with the ORR following anti-PD1 monotherapy reported in the literature. Lower correlations were found with other immune-related genes/signatures, including PDL1. Using the same population-based cutoff (80th percentile), similar proportions of PD1-high disease in a given cancer type were identified in our in-house 773 tumor dataset as compared with TCGA. Finally, the pre-established PD1 mRNA FFPE-based cutoff was found significantly associated with anti-PD1 response in 117 patients with advanced disease (PD1-high 51.5%, PD1-intermediate 26.6% and PD1-low 15.0%; odds ratio between PD1-high and PD1-intermediate/low = 8.31; P < 0.001). In this same dataset, PDL1 tumor expression by IHC or percentage of sTILs was not found associated with response., Conclusions: Our study provides a clinically applicable assay that links PD1 mRNA abundance, activated CD8 T-cells and anti-PD1 efficacy.
- Published
- 2018
- Full Text
- View/download PDF
34. Liquid biopsies come of age: towards implementation of circulating tumour DNA.
- Author
-
Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, Pacey S, Baird R, and Rosenfeld N
- Subjects
- Biomarkers, Tumor blood, Clonal Evolution, Early Detection of Cancer, ErbB Receptors genetics, Humans, Mutation, Neoplasm Staging, Neoplasms genetics, Precision Medicine, Biopsy, DNA, Neoplasm blood, Neoplasms pathology
- Abstract
Improvements in genomic and molecular methods are expanding the range of potential applications for circulating tumour DNA (ctDNA), both in a research setting and as a 'liquid biopsy' for cancer management. Proof-of-principle studies have demonstrated the translational potential of ctDNA for prognostication, molecular profiling and monitoring. The field is now in an exciting transitional period in which ctDNA analysis is beginning to be applied clinically, although there is still much to learn about the biology of cell-free DNA. This is an opportune time to appraise potential approaches to ctDNA analysis, and to consider their applications in personalized oncology and in cancer research.
- Published
- 2017
- Full Text
- View/download PDF
35. A phase I pharmacokinetic and safety study of cabazitaxel in adult cancer patients with normal and impaired renal function.
- Author
-
Azaro A, Rodón J, Machiels JP, Rottey S, Damian S, Baird R, Garcia-Corbacho J, Mathijssen RHJ, Clot PF, Wack C, Shen L, and de Jonge MJA
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Female, Humans, Male, Middle Aged, Neoplasms metabolism, Taxoids adverse effects, Antineoplastic Agents pharmacokinetics, Neoplasms drug therapy, Renal Insufficiency metabolism, Taxoids pharmacokinetics
- Abstract
Purpose: Limited data are available on cabazitaxel pharmacokinetics in patients with renal impairment. This open-label, multicenter study assessed cabazitaxel in patients with advanced solid tumors and normal or impaired renal function., Methods: Cohorts A (normal renal function: creatinine clearance [CrCL] >80 mL/min/1.73 m
2 ), B (moderate renal impairment: CrCL 30 to <50 mL/min/1.73 m2 ) and C (severe impairment: CrCL <30 mL/min/1.73 m2 ) received cabazitaxel 25 mg/m2 (A, B) or 20 mg/m2 (C, could be escalated to 25 mg/m2 ), once every 3 weeks. Pharmacokinetic parameters and cabazitaxel unbound fraction (FU ) were assessed using linear regression and mixed models. Geometric mean (GM) and GM ratios (GMRs) were determined using mean CrCL intervals (moderate and severe renal impairment: 40 and 15 mL/min/1.73 m2 ) versus a control (90 mL/min/1.73 m2 )., Results: Overall, 25 patients received cabazitaxel (median cycles: 3 [range 1-20]; Cohort A: 5 [2-13]; Cohort B: 3 [1-15]; and Cohort C: 5 [1-20]), of which 24 were eligible for pharmacokinetic analysis (eight in each cohort). For moderate and severe renal impairment versus normal renal function, GMR estimates were: clearance normalized to body surface area (CL/BSA) 0.95 (90% CI 0.80-1.13) and 0.89 (0.61-1.32); area under the curve normalized to dose (AUC/dose) 1.06 (0.88-1.27) and 1.14 (0.76-1.71); and FU 0.99 (0.94-1.04) and 0.97 (0.87-1.09), respectively. Estimated slopes of linear regression of log parameters versus log CrCL (renal impairment) were: CL/BSA 0.06 (-0.15 to 0.28); AUC/dose -0.07 (-0.30 to 0.16); and FU 0.02 (-0.05 to 0.08). Cabazitaxel safety profile was consistent with previous reports., Conclusions: Renal impairment had no clinically meaningful effect on cabazitaxel pharmacokinetics., Competing Interests: Compliance with ethical standardsConflict of interestThis study was sponsored by Sanofi. Analía Azaro, Jordi Rodón, Silvia Damian, Javier Garcia-Corbacho and Maja de Jonge have no conflicts of interest to disclose. Jean-Pascal Machiels has been a member of advisory boards for Boehringer Ingelheim (without compensation) and MSD, and received research grants from Novartis, Bayer and Janssen. Sylvie Rottey has been a member of advisory boards and received research funding from Sanofi. Richard Baird and Ron Mathijssen have received research funding from Sanofi. Pierre-François Clot, Claudine Wack, and Liji Shen are employees of Sanofi. Liji Shen is a stock holder of Sanofi.Ethical approvalAll procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.