Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where C. Hudis is active.

Publication


Featured researches published by C. Hudis.


Journal of Clinical Oncology | 1996

Phase II study of weekly intravenous recombinant humanized anti-p185HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer.

José Baselga; Debasish Tripathy; John Mendelsohn; S. Baughman; Christopher C. Benz; L. Dantis; N. T. Sklarin; A. D. Seidman; C. Hudis; J. Moore; P. P. Rosen; T. Twaddell; I. C. Henderson; L. Norton

PURPOSE Breast cancer frequently overexpresses the product of the HER2 proto-oncogene, a 185-kd growth factor receptor (p185HER2). The recombinant humanized monoclonal antibody (rhuMAb) HER2 has high affinity for p185HER2 and inhibits the growth of breast cancer cells that overexpress HER2. We evaluated the efficacy and toxicity of weekly intravenous administration of rhuMAb HER2 in patients with HER2-overexpressing metastatic breast cancer. PATIENTS AND METHODS We treated 46 patients with metastatic breast carcinomas that overexpressed HER2. Patients received a loading dose of 250 mg of intravenous rhuMAb HER2, then 10 weekly doses of 100 mg each. Patients with no disease progression at the completion of this treatment period were offered a maintenance phase of 100 mg/wk. RESULTS Study patients had extensive metastatic disease, and most had received extensive prior anticancer therapy. Adequate pharmacokinetic levels of rhuMAb HER2 were obtained in 90% of the patients. Toxicity was minimal and no antibodies against rhuMAb HER2 were detected in any patients. Objective responses were seen in five of 43 assessable patients, and included one complete remission and four partial remissions (overall response rate, 11.6%; 95% confidence interval, 4.36 to 25.9). Responses were observed in liver, mediastinum, lymph nodes, and chest wall lesions. Minor responses, seen in two patients, and stable disease, which occurred in 14 patients, lasted for a median of 5.1 months. CONCLUSION rhuMAb HER2 is well tolerated and clinically active in patients with HER2-overexpressing metastatic breast cancers that had received extensive prior therapy. This is evidence that targeting growth factor receptors can cause regression of human cancer and justifies further evaluation of this agent.


Annals of Oncology | 2014

ESO-ESMO 2nd international consensus guidelines for advanced breast cancer (ABC2)†

Fatima Cardoso; Alberto Costa; Larry Norton; Elżbieta Senkus; M. Aapro; Fabrice Andre; Carlos H. Barrios; Jonas Bergh; L. Biganzoli; Kimberly L. Blackwell; Maria João Cardoso; Tanja Cufer; N. El Saghir; Lesley Fallowfield; D. Fenech; Prudence A. Francis; K. Gelmon; Sharon H. Giordano; Joseph Gligorov; A. Goldhirsch; Nadia Harbeck; Nehmat Houssami; C. Hudis; Bella Kaufman; Ian E. Krop; Stella Kyriakides; U.N. Lin; Musa Mayer; S.D. Merjaver; E.B. Nordström

Advanced Breast Cancer (ABC) is a treatable but still generally incurable disease. The goals of care are to optimize both length and quality of life. Due to continuous research, several advances have been made, particularly for the HER-2-positive and for Luminal-like subtypes. Notwithstanding these advances, median overall survival of patients with ABC is still only 2–3 years, although the range is wide [1–5], and survival may be longer for patients treated in specialized institutions [6]. Implementation of current knowledge is highly variable among countries and within each country.


Journal of Clinical Oncology | 2012

Pathologic Complete Response Predicts Recurrence-Free Survival More Effectively by Cancer Subset: Results From the I-SPY 1 TRIAL—CALGB 150007/150012, ACRIN 6657

Laura Esserman; Donald A. Berry; Angela DeMichele; Lisa A. Carey; Sarah E. Davis; Meredith Buxton; C. Hudis; Joe W. Gray; Charles M. Perou; Christina Yau; Chad A. Livasy; Helen Krontiras; Leslie Montgomery; Debasish Tripathy; Constance D. Lehman; Minetta C. Liu; Olufunmilayo I. Olopade; Hope S. Rugo; John T. Carpenter; Lynn G. Dressler; David C. Chhieng; Baljit Singh; Carolyn Mies; Joseph T. Rabban; Yunn-Yi Chen; Dilip Giri; Laura J. van 't Veer; Nola M. Hylton

PURPOSE Neoadjuvant chemotherapy for breast cancer provides critical information about tumor response; how best to leverage this for predicting recurrence-free survival (RFS) is not established. The I-SPY 1 TRIAL (Investigation of Serial Studies to Predict Your Therapeutic Response With Imaging and Molecular Analysis) was a multicenter breast cancer study integrating clinical, imaging, and genomic data to evaluate pathologic response, RFS, and their relationship and predictability based on tumor biomarkers. PATIENTS AND METHODS Eligible patients had tumors ≥ 3 cm and received neoadjuvant chemotherapy. We determined associations between pathologic complete response (pCR; defined as the absence of invasive cancer in breast and nodes) and RFS, overall and within receptor subsets. RESULTS In 221 evaluable patients (median tumor size, 6.0 cm; median age, 49 years; 91% classified as poor risk on the basis of the 70-gene prognosis profile), 41% were hormone receptor (HR) negative, and 31% were human epidermal growth factor receptor 2 (HER2) positive. For 190 patients treated without neoadjuvant trastuzumab, pCR was highest for HR-negative/HER2-positive patients (45%) and lowest for HR-positive/HER2-negative patients (9%). Achieving pCR predicted favorable RFS. For 172 patients treated without trastuzumab, the hazard ratio for RFS of pCR versus no pCR was 0.29 (95% CI, 0.07 to 0.82). pCR was more predictive of RFS by multivariate analysis when subtype was taken into account, and point estimates of hazard ratios within the HR-positive/HER2-negative (hazard ratio, 0.00; 95% CI, 0.00 to 0.93), HR-negative/HER2-negative (hazard ratio, 0.25; 95% CI, 0.04 to 0.97), and HER2-positive (hazard ratio, 0.14; 95% CI, 0.01 to 1.0) subtypes are lower. Ki67 further improved the prediction of pCR within subsets. CONCLUSION In this biologically high-risk group, pCR differs by receptor subset. pCR is more highly predictive of RFS within every established receptor subset than overall, demonstrating that the extent of outcome advantage conferred by pCR is specific to tumor biology.


The Breast | 2017

3rd ESO-ESMO international consensus guidelines for Advanced Breast Cancer (ABC 3).

Fatima Cardoso; Ana Costa; Elżbieta Senkus; M. Aapro; Fabrice Andre; Carlos H. Barrios; Jonas Bergh; G. Bhattacharyya; Laura Biganzoli; Maria João Cardoso; Lisa A. Carey; D. Corneliussen-James; Giuseppe Curigliano; V. Dieras; N. El Saghir; Alexandru Eniu; Lesley Fallowfield; D. Fenech; Prudence A. Francis; Karen A. Gelmon; A. Gennari; Nadia Harbeck; C. Hudis; Bella Kaufman; Ian E. Krop; Musa Mayer; H. Meijer; S. Mertz; Shinji Ohno; Olivia Pagani

• This ESO-ESMO ABC 5 Clinical Practice Guideline provides key recommendations for managing advanced breast cancer patients. • It provides updates on managing patients with all breast cancer subtypes, LABC, follow-up, palliative and supportive care. • Updated diagnostic and treatment algorithms are also provided. • All recommendations were compiled by a multidisciplinary group of international experts. • Recommendations are based on available clinical evidence and the collective expert opinion of the authors.


British Journal of Cancer | 2014

Predicting response and survival in chemotherapy-treated triple-negative breast cancer

Aleix Prat; Ana Lluch; Joan Albanell; William T. Barry; C Fan; J I Chacón; Joel S. Parker; L Calvo; A Plazaola; A Arcusa; M A Seguí-Palmer; O Burgues; N Ribelles; Álvaro Rodríguez-Lescure; A Guerrero; Manuel Ruiz-Borrego; Blanca Munárriz; J A López; B Adamo; Maggie Cheang; Y Li; Zhiyuan Hu; M L Gulley; M J Vidal; Brandy Pitcher; Minetta C. Liu; Marc L. Citron; Matthew J. Ellis; Elaine R. Mardis; Tammi L. Vickery

Background:In this study, we evaluated the ability of gene expression profiles to predict chemotherapy response and survival in triple-negative breast cancer (TNBC).Methods:Gene expression and clinical–pathological data were evaluated in five independent cohorts, including three randomised clinical trials for a total of 1055 patients with TNBC, basal-like disease (BLBC) or both. Previously defined intrinsic molecular subtype and a proliferation signature were determined and tested. Each signature was tested using multivariable logistic regression models (for pCR (pathological complete response)) and Cox models (for survival). Within TNBC, interactions between each signature and the basal-like subtype (vs other subtypes) for predicting either pCR or survival were investigated.Results:Within TNBC, all intrinsic subtypes were identified but BLBC predominated (55–81%). Significant associations between genomic signatures and response and survival after chemotherapy were only identified within BLBC and not within TNBC as a whole. In particular, high expression of a previously identified proliferation signature, or low expression of the luminal A signature, was found independently associated with pCR and improved survival following chemotherapy across different cohorts. Significant interaction tests were only obtained between each signature and the BLBC subtype for prediction of chemotherapy response or survival.Conclusions:The proliferation signature predicts response and improved survival after chemotherapy, but only within BLBC. This highlights the clinical implications of TNBC heterogeneity, and suggests that future clinical trials focused on this phenotypic subtype should consider stratifying patients as having BLBC or not.


Clinical Cancer Research | 2013

Sorafenib or Placebo with Either Gemcitabine or Capecitabine in Patients with HER-2–Negative Advanced Breast Cancer That Progressed during or after Bevacizumab

Lee S. Schwartzberg; Kw Tauer; Rc Hermann; Grace Makari-Judson; Claudine Isaacs; Jt Beck; Kaklamani; Ej Stepanski; Hope S. Rugo; W Wang; Katherine M. Bell-McGuinn; Jeffrey J. Kirshner; Peter D. Eisenberg; R Emanuelson; M Keaton; Ellis G. Levine; Dc Medgyesy; R Qamar; A Starr; Sk Ro; Nathalie Andrienne Lokker; C. Hudis

Purpose: We assessed adding the multikinase inhibitor sorafenib to gemcitabine or capecitabine in patients with advanced breast cancer whose disease progressed during/after bevacizumab. Experimental Design: This double-blind, randomized, placebo-controlled phase IIb study (ClinicalTrials.gov NCT00493636) enrolled patients with locally advanced or metastatic human epidermal growth factor receptor 2 (HER2)–negative breast cancer and prior bevacizumab treatment. Patients were randomized to chemotherapy with sorafenib (400 mg, twice daily) or matching placebo. Initially, chemotherapy was gemcitabine (1,000 mg/m2 i.v., days 1, 8/21), but later, capecitabine (1,000 mg/m2 orally twice daily, days 1–14/21) was allowed as an alternative. The primary endpoint was progression-free survival (PFS). Results: One hundred and sixty patients were randomized. More patients received gemcitabine (82.5%) than capecitabine (17.5%). Sorafenib plus gemcitabine/capecitabine was associated with a statistically significant prolongation in PFS versus placebo plus gemcitabine/capecitabine [3.4 vs. 2.7 months; HR = 0.65; 95% confidence interval (CI): 0.45–0.95; P = 0.02], time to progression was increased (median, 3.6 vs. 2.7 months; HR = 0.64; 95% CI: 0.44–0.93; P = 0.02), and overall response rate was 19.8% versus 12.7% (P = 0.23). Median survival was 13.4 versus 11.4 months for sorafenib versus placebo (HR = 1.01; 95% CI: 0.71−1.44; P = 0.95). Addition of sorafenib versus placebo increased grade 3/4 hand–foot skin reaction (39% vs. 5%), stomatitis (10% vs. 0%), fatigue (18% vs. 9%), and dose reductions that were more frequent (51.9% vs. 7.8%). Conclusion: The addition of sorafenib to gemcitabine/capecitabine provided a clinically small but statistically significant PFS benefit in HER2-negative advanced breast cancer patients whose disease progressed during/after bevacizumab. Combination treatment was associated with manageable toxicities but frequently required dose reductions. Clin Cancer Res; 19(10); 2745–54. ©2013 AACR.


Annals of Oncology | 2014

Phase III trial of nonpegylated liposomal doxorubicin in combination with trastuzumab and paclitaxel in HER2-positive metastatic breast cancer

José Baselga; A. Manikhas; J. Cortés; A. Llombart; Laslo Roman; Vladimir Semiglazov; M. Byakhov; D. Lokanatha; S. Forenza; R. H. Goldfarb; J. Matera; N. Azarnia; C. Hudis; M. Rozencweig

BACKGROUND Nonpegylated liposomal doxorubicin liposomal doxorubicin, (Myocet™; Sopherion Therapeutics, Inc Canada, and Cephalon, Europe) (NPLD; Myocet(®)) in combination with trastuzumabHerceptin(®) (Hoffmann-La Roche) has shown promising activity and cardiac safety. We conducted a randomized phase III trial of first-line NPLD plus trastuzumab and paclitaxel (Pharmachemie B.V.) (MTP) versus trastuzumab plus paclitaxel (TP) in patients with human epidermal growth factor 2 receptor (HER2)-positive metastatic breast cancer. PATIENTS AND METHODS Patients were randomly assigned to NPLD (M, 50 mg/m(2) every 3 weeks for six cycles), trastuzumab (T, 4 mg/kg loading dose followed by 2 mg/kg weekly), and paclitaxel (P, 80 mg/m(2) weekly) or T + P at the same doses until progression or toxicity. The primary efficacy outcome was progression-free survival (PFS). RESULTS One hundred and eighty-one patients were allocated to receive MTP, and 183 to TP. Median PFS was 16.1 and 14.5 months with MTP and TP, respectively [hazard ratio (HR) 0.84; two-sided P = 0.174]. In patients with estrogen receptor (ER)- and progesterone receptor (PR)-negative tumors, PFS was 20.7 and 14.0 months, respectively [HR 0.68; 95% confidence interval (CI) 0.47-0.99]. Median overall survival (OS) was 33.6 and 28.9 months with MTP and TP, respectively (HR 0.79; two-sided P = 0.083). In ER- and PR-negative tumors, OS was 38.2 and 27.9 months, respectively (HR 0.63; 95% CI 0.42-0.93). The frequency of adverse events was higher with MTP, but there was no significant difference in cardiac toxicity between treatment arms. CONCLUSION(S) The trial failed to demonstrate a significant clinical improvement with the addition of M to TP regimen. The clinical benefit observed in an exploratory analysis in the ER- and PR-negative population deserves consideration for further clinical trials. CLINICAL TRIAL NUMBER NCT00294996.BACKGROUND Nonpegylated liposomal doxorubicin liposomal doxorubicin, (Myocet™; Sopherion Therapeutics, Inc Canada, and Cephalon, Europe) (NPLD; Myocet®) in combination with trastuzumabHerceptin® (Hoffmann-La Roche) has shown promising activity and cardiac safety. We conducted a randomized phase III trial of first-line NPLD plus trastuzumab and paclitaxel (Pharmachemie B.V.) (MTP) versus trastuzumab plus paclitaxel (TP) in patients with human epidermal growth factor 2 receptor (HER2)-positive metastatic breast cancer. PATIENTS AND METHODS Patients were randomly assigned to NPLD (M, 50 mg/m2 every 3 weeks for six cycles), trastuzumab (T, 4 mg/kg loading dose followed by 2 mg/kg weekly), and paclitaxel (P, 80 mg/m2 weekly) or T + P at the same doses until progression or toxicity. The primary efficacy outcome was progression-free survival (PFS). RESULTS One hundred and eighty-one patients were allocated to receive MTP, and 183 to TP. Median PFS was 16.1 and 14.5 months with MTP and TP, respectively [hazard ratio (HR) 0.84; two-sided P = 0.174]. In patients with estrogen receptor (ER)- and progesterone receptor (PR)-negative tumors, PFS was 20.7 and 14.0 months, respectively [HR 0.68; 95% confidence interval (CI) 0.47-0.99]. Median overall survival (OS) was 33.6 and 28.9 months with MTP and TP, respectively (HR 0.79; two-sided P = 0.083). In ER- and PR-negative tumors, OS was 38.2 and 27.9 months, respectively (HR 0.63; 95% CI 0.42-0.93). The frequency of adverse events was higher with MTP, but there was no significant difference in cardiac toxicity between treatment arms. CONCLUSION(S) The trial failed to demonstrate a significant clinical improvement with the addition of M to TP regimen. The clinical benefit observed in an exploratory analysis in the ER- and PR-negative population deserves consideration for further clinical trials. CLINICAL TRIAL NUMBER NCT00294996.


Annals of Oncology | 2011

A phase I study of dasatinib and weekly paclitaxel for metastatic breast cancer

Monica Fornier; Patrick G. Morris; A. Abbruzzi; G. D'andrea; T. Gilewski; J. Bromberg; Chau Dang; Maura N. Dickler; S. Modi; Andrew D. Seidman; N. T. Sklarin; Jenny Chang; Larry Norton; C. Hudis

BACKGROUND SRC plays an important role in the pathogenesis of metastatic breast cancer (MBC). In preclinical models, paclitaxel and the oral SRC inhibitor dasatinib showed greater antitumor activity than either agent. To determine the maximum tolerated dose of this combination, we conducted a phase I study. PATIENTS AND METHODS Patients with MBC; Eastern Cooperative Oncology Group performance status of zero to one; normal hepatic, renal and marrow function were eligible. Paclitaxel 80 mg/m(2) was given 3 weeks of 4. The starting dasatinib dose was 70 mg and was increased, using a standard 3 + 3 dose-escalation scheme. RESULTS Fifteen patients enrolled (median age 54 years, range 35-74). No dose-limiting toxic effects (DLTs) occurred at dasatinib doses of 70-120 mg. One DLT (grade 3 fatigue) occurred in the dasatinib 150-mg cohort, which was expanded (six patients) with no further DLTs. However, due to cumulative toxic effects (rash, fatigue, diarrhea), the recommended phase II dose is dasatinib 120 mg. Of 13 assessable patients, a partial response was seen in 4 patients (31%), including 2 patients previously treated with taxanes; all received ≥120 mg dasatinib. An additional five patients (29%) had stable disease. CONCLUSION In combination with weekly paclitaxel, the recommended phase II dose of dasatinib is 120 mg daily and preliminary activity has been seen in patients with MBC.


Lancet Oncology | 2017

Future cancer research priorities in the USA: a Lancet Oncology Commission

Elizabeth M. Jaffee; Chi Van Dang; David B. Agus; Brian M. Alexander; Kenneth C. Anderson; Alan Ashworth; Anna Barker; Roshan Bastani; Sangeeta N. Bhatia; Jeffrey A. Bluestone; Otis W. Brawley; Atul J. Butte; Daniel G. Coit; Nancy E. Davidson; Mark E. Davis; Ronald A. DePinho; Robert B. Diasio; Giulio Draetta; A. Lindsay Frazier; Andrew Futreal; S. S. Gambhir; Patricia A. Ganz; Levi A. Garraway; Stanton L. Gerson; Sumit Gupta; James R. Heath; Ruth I. Hoffman; C. Hudis; Chanita Hughes-Halbert; Ramy Ibrahim

We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US


Molecular Oncology | 2015

Serum metabolomic profiles evaluated after surgery may identify patients with oestrogen receptor negative early breast cancer at increased risk of disease recurrence. Results from a retrospective study.

Leonardo Tenori; Catherine Oakman; Patrick G. Morris; Ewa Gralka; Natalie Turner; Silvia Cappadona; Monica Fornier; C. Hudis; Larry Norton; Claudio Luchinat; Angelo Di Leo

2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.

Collaboration


Dive into the C. Hudis's collaboration.

Top Co-Authors

Avatar

Sujata Patil

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Larry Norton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tiffany A. Traina

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maura N. Dickler

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ayca Gucalp

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chau T. Dang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Andrew D. Seidman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shari Goldfarb

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge