Network


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

Hotspot


Dive into the research topics where Richard D. Baird is active.

Publication


Featured researches published by Richard D. Baird.


Journal of Clinical Oncology | 2011

First-in-Man Clinical Trial of the Oral Pan-AKT Inhibitor MK-2206 in Patients With Advanced Solid Tumors

Timothy A. Yap; Li Yan; Amita Patnaik; Ivy Fearen; David Olmos; Kyriakos P. Papadopoulos; Richard D. Baird; Liliana Delgado; Adekemi Taylor; Lisa Lupinacci; Ruth Riisnaes; Lorna Pope; Simon P. Heaton; George Thomas; Michelle D. Garrett; Daniel M. Sullivan; Johann S. de Bono; Anthony W. Tolcher

PURPOSE AKT signaling is frequently deregulated in human cancers. MK-2206 is a potent, oral allosteric inhibitor of all AKT isoforms with antitumor activity in preclinical models. A phase I study of MK-2206 was conducted to investigate its safety, maximum-tolerated dose (MTD), pharmacokinetics (PKs), pharmacodynamics (PDs), and preliminary antitumor efficacy. PATIENTS AND METHODS Patients with advanced solid tumors received MK-2206 on alternate days. Paired tumor biopsies were mandated at the MTD for biomarker studies. PD studies incorporated tumor and hair follicle analyses, and putative predictive biomarker studies included tumor somatic mutation analyses and immunohistochemistry for phosphatase and tensin homolog (PTEN) loss. RESULTS Thirty-three patients received MK-2206 at 30, 60, 75, or 90 mg on alternate days. Dose-limiting toxicities included skin rash and stomatitis, establishing the MTD at 60 mg. Drug-related toxicities included skin rash (51.5%), nausea (36.4%), pruritus (24.2%), hyperglycemia (21.2%), and diarrhea (21.2%). PKs (area under the concentration-time curve from 0 to 48 hours and maximum measured plasma concentration) were dose proportional. Phosphorylated serine 473 AKT declined in all tumor biopsies assessed (P = .015), and phosphorylated threonine 246 proline-rich AKT substrate 40 was suppressed in hair follicles at 6 hours (P = .008), on days 7 (P = .028) and 15 (P = .025) with MK-2206; reversible hyperglycemia and increases in insulin c-peptide were also observed, confirming target modulation. A patient with pancreatic adenocarcinoma (PTEN loss; KRAS G12D mutation) treated at 60 mg on alternate days experienced a decrease of approximately 60% in cancer antigen 19-9 levels and 23% shrinkage in tumor measurements. Two patients with pancreatic neuroendocrine tumors had minor tumor responses. CONCLUSION MK-2206 was well tolerated, with evidence of AKT signaling blockade. Rational combination trials are ongoing to maximize clinical benefit with this therapeutic strategy.


Lancet Oncology | 2013

The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: A phase 1 dose-escalation trial

Shahneen Sandhu; William R. Schelman; George Wilding; Victor Moreno; Richard D. Baird; Susana Miranda; Lucy Hylands; Ruth Riisnaes; Martin Forster; Aurelius Omlin; Nathan Kreischer; Khin Thway; Heidrun Gevensleben; Linda Sun; John W. Loughney; Manash Shankar Chatterjee; Carlo Toniatti; Christopher L. Carpenter; Robert Iannone; Stan B. Kaye; Johann S. de Bono; Robert M Wenham

BACKGROUND Poly(ADP-ribose) polymerase (PARP) is implicated in DNA repair and transcription regulation. Niraparib (MK4827) is an oral potent, selective PARP-1 and PARP-2 inhibitor that induces synthetic lethality in preclinical tumour models with loss of BRCA and PTEN function. We investigated the safety, tolerability, maximum tolerated dose, pharmacokinetic and pharmacodynamic profiles, and preliminary antitumour activity of niraparib. METHODS In a phase 1 dose-escalation study, we enrolled patients with advanced solid tumours at one site in the UK and two sites in the USA. Eligible patients were aged at least 18 years; had a life expectancy of at least 12 weeks; had an Eastern Cooperative Oncology Group performance status of 2 or less; had assessable disease; were not suitable to receive any established treatments; had adequate organ function; and had discontinued any previous anticancer treatments at least 4 weeks previously. In part A, cohorts of three to six patients, enriched for BRCA1 and BRCA2 mutation carriers, received niraparib daily at ten escalating doses from 30 mg to 400 mg in a 21-day cycle to establish the maximum tolerated dose. Dose expansion at the maximum tolerated dose was pursued in 15 patients to confirm tolerability. In part B, we further investigated the maximum tolerated dose in patients with sporadic platinum-resistant high-grade serous ovarian cancer and sporadic prostate cancer. We obtained blood, circulating tumour cells, and optional paired tumour biopsies for pharmacokinetic and pharmacodynamic assessments. Toxic effects were assessed by common toxicity criteria and tumour responses ascribed by Response Evaluation Criteria in Solid Tumors (RECIST). Circulating tumour cells and archival tumour tissue in prostate patients were analysed for exploratory putative predictive biomarkers, such as loss of PTEN expression and ETS rearrangements. This trial is registered with ClinicalTrials.gov, NCT00749502. FINDINGS Between Sept 15, 2008, and Jan 14, 2011, we enrolled 100 patients: 60 in part A and 40 in part B. 300 mg/day was established as the maximum tolerated dose. Dose-limiting toxic effects reported in the first cycle were grade 3 fatigue (one patient given 30 mg/day), grade 3 pneumonitis (one given 60 mg/day), and grade 4 thrombocytopenia (two given 400 mg/day). Common treatment-related toxic effects were anaemia (48 patients [48%]), nausea (42 [42%]), fatigue (42 [42%]), thrombocytopenia (35 [35%]), anorexia (26 [26%]), neutropenia (24 [24%]), constipation (23 [23%]), and vomiting (20 [20%]), and were predominantly grade 1 or 2. Pharmacokinetics were dose proportional and the mean terminal elimination half-life was 36·4 h (range 32·8-46·0). Pharmacodynamic analyses confirmed PARP inhibition exceeded 50% at doses greater than 80 mg/day and antitumour activity was documented beyond doses of 60 mg/day. Eight (40% [95% CI 19-64]) of 20 BRCA1 or BRCA2 mutation carriers with ovarian cancer had RECIST partial responses, as did two (50% [7-93]) of four mutation carriers with breast cancer. Antitumour activity was also reported in sporadic high-grade serous ovarian cancer, non-small-cell lung cancer, and prostate cancer. We recorded no correlation between loss of PTEN expression or ETS rearrangements and measures of antitumour activity in patients with prostate cancer. INTERPRETATION A recommended phase 2 dose of 300 mg/day niraparib is well tolerated. Niraparib should be further assessed in inherited and sporadic cancers with homologous recombination DNA repair defects and to target PARP-mediated transcription in cancer. FUNDING Merck Sharp and Dohme.


European Journal of Cancer | 2003

Drug resistance reversal--are we getting closer?

Richard D. Baird; Stan B. Kaye

Clinical drug resistance is a major barrier to overcome before chemotherapy can become curative for most patients presenting with metastatic cancer. Rational attempts to tackle clinical drug resistance need to be based on an understanding of the mechanisms involved; these are likely to be complex and multifactorial, and may be due to inadequate drug exposure or alterations in the cancer cell itself. This article reviews a number of strategies used to tackle drug resistance, focussing on work in our institution related to the treatment of ovarian cancer and resistance to platinum and taxane-based chemotherapy. Further progress towards drug resistance reversal will require a three-pronged approach, namely: the development of novel cytotoxics which exploit selectively expressed targets; modulation of resistance to conventional agents and, most importantly, a serious attempt to understand resistance mechanisms in tumour samples taken both pre- and post-chemotherapy.


Clinical Cancer Research | 2015

First-in-Human Phase I Study of Pictilisib (GDC-0941), a Potent Pan–Class I Phosphatidylinositol-3-Kinase (PI3K) Inhibitor, in Patients with Advanced Solid Tumors

Debashis Sarker; Joo Ern Ang; Richard D. Baird; Rebecca Kristeleit; K. Shah; Victor Moreno; Paul A. Clarke; Florence I. Raynaud; Gallia G. Levy; Joseph A. Ware; K. E. Mazina; Ray S. Lin; Jenny Wu; Jill Fredrickson; Jill M. Spoerke; Mark R. Lackner; Yibing Yan; Lori S. Friedman; Stan B. Kaye; Mika K. Derynck; Paul Workman; Johann S. de Bono

Purpose: This first-in-human dose-escalation trial evaluated the safety, tolerability, maximal-tolerated dose (MTD), dose-limiting toxicities (DLT), pharmacokinetics, pharmacodynamics, and preliminary clinical activity of pictilisib (GDC-0941), an oral, potent, and selective inhibitor of the class I phosphatidylinositol-3-kinases (PI3K). Patients and Methods: Sixty patients with solid tumors received pictilisib at 14 dose levels from 15 to 450 mg once-daily, initially on days 1 to 21 every 28 days and later, using continuous dosing for selected dose levels. Pharmacodynamic studies incorporated 18F-FDG-PET, and assessment of phosphorylated AKT and S6 ribosomal protein in platelet-rich plasma (PRP) and tumor tissue. Results: Pictilisib was well tolerated. The most common toxicities were grade 1–2 nausea, rash, and fatigue, whereas the DLT was grade 3 maculopapular rash (450 mg, 2 of 3 patients; 330 mg, 1 of 7 patients). The pharmacokinetic profile was dose-proportional and supported once-daily dosing. Levels of phosphorylated serine-473 AKT were suppressed >90% in PRP at 3 hours after dose at the MTD and in tumor at pictilisib doses associated with AUC >20 h·μmol/L. Significant increase in plasma insulin and glucose levels, and >25% decrease in 18F-FDG uptake by PET in 7 of 32 evaluable patients confirmed target modulation. A patient with V600E BRAF–mutant melanoma and another with platinum-refractory epithelial ovarian cancer exhibiting PTEN loss and PIK3CA amplification demonstrated partial response by RECIST and GCIG-CA125 criteria, respectively. Conclusion: Pictilisib was safely administered with a dose-proportional pharmacokinetic profile, on-target pharmacodynamic activity at dose levels ≥100 mg and signs of antitumor activity. The recommended phase II dose was continuous dosing at 330 mg once-daily. Clin Cancer Res; 21(1); 77–86. ©2014 AACR.


Cell | 2016

A Biobank of Breast Cancer Explants with Preserved Intra-tumor Heterogeneity to Screen Anticancer Compounds

Alejandra Bruna; Oscar M. Rueda; Wendy Greenwood; Ankita Sati Batra; Maurizio Callari; R.N. Batra; Katherine Pogrebniak; Jose L. Sandoval; John W Cassidy; Ana Tufegdzic-Vidakovic; Stephen John Sammut; Linda Jones; Elena Provenzano; Richard D. Baird; Peter Eirew; James Hadfield; Matthew Eldridge; Anne McLaren-Douglas; Andrew Barthorpe; Howard Lightfoot; Mark J. O’Connor; Joe W. Gray; Javier Cortes; José Baselga; Elisabetta Marangoni; Alana L. Welm; Samuel Aparicio; Violeta Serra; Mathew J. Garnett; Carlos Caldas

Summary The inter- and intra-tumor heterogeneity of breast cancer needs to be adequately captured in pre-clinical models. We have created a large collection of breast cancer patient-derived tumor xenografts (PDTXs), in which the morphological and molecular characteristics of the originating tumor are preserved through passaging in the mouse. An integrated platform combining in vivo maintenance of these PDTXs along with short-term cultures of PDTX-derived tumor cells (PDTCs) was optimized. Remarkably, the intra-tumor genomic clonal architecture present in the originating breast cancers was mostly preserved upon serial passaging in xenografts and in short-term cultured PDTCs. We assessed drug responses in PDTCs on a high-throughput platform and validated several ex vivo responses in vivo. The biobank represents a powerful resource for pre-clinical breast cancer pharmacogenomic studies (http://caldaslab.cruk.cam.ac.uk/bcape), including identification of biomarkers of response or resistance.


Clinical Cancer Research | 2015

Antitumor activity in ras-driven tumors by blocking akt and mek

Anthony W. Tolcher; Khurum Khan; Michael Ong; Udai Banerji; Vassiliki Papadimitrakopoulou; David R. Gandara; Amita Patnaik; Richard D. Baird; David Olmos; Christopher R. Garrett; Jeffrey M. Skolnik; Eric H. Rubin; Paul D. Smith; Pearl S. Huang; Maria Learoyd; Keith Shannon; Anne Morosky; Ernestina Tetteh; Ying Ming Jou; Kyriakos P. Papadopoulos; Victor Moreno; Brianne Kaiser; Timothy A. Yap; Li Yan; Johann S. de Bono

Purpose: KRAS is the most commonly mutated oncogene in human tumors. KRAS-mutant cells may exhibit resistance to the allosteric MEK1/2 inhibitor selumetinib (AZD6244; ARRY-142886) and allosteric AKT inhibitors (such as MK-2206), the combination of which may overcome resistance to both monotherapies. Experimental Design: We conducted a dose/schedule-finding study evaluating MK-2206 and selumetinib in patients with advanced treatment-refractory solid tumors. Recommended dosing schedules were defined as MK-2206 at 135 mg weekly and selumetinib at 100 mg once daily. Results: Grade 3 rash was the most common dose-limiting toxicity (DLT); other DLTs included grade 4 lipase increase, grade 3 stomatitis, diarrhea, and fatigue, and grade 3 and grade 2 retinal pigment epithelium detachment. There were no meaningful pharmacokinetic drug–drug interactions. Clinical antitumor activity included RECIST 1.0–confirmed partial responses in non–small cell lung cancer and low-grade ovarian carcinoma. Conclusion: Responses in KRAS-mutant cancers were generally durable. Clinical cotargeting of MEK and AKT signaling may be an important therapeutic strategy in KRAS-driven human malignancies (Trial NCT number NCT01021748). Clin Cancer Res; 21(4); 739–48. ©2014 AACR.


Nature Reviews Clinical Oncology | 2010

Weekly paclitaxel in the treatment of recurrent ovarian cancer

Richard D. Baird; David Sp Tan; Stan B. Kaye

Weekly paclitaxel is a highly active and well tolerated regimen that is increasingly being adopted for the treatment of relapsed ovarian cancer. This regimen is usually administered at 80–90 mg/m2/week, and the use of a 1 h infusion helps minimize myelosuppression. When compared with the 3-weekly schedule, weekly paclitaxel is better tolerated, with a reduced frequency of grade 3–4 toxic effects. Single-agent weekly paclitaxel for relapsed ovarian cancer yields response rates in the range of 20–62%; however, response duration can be short. Responses to weekly paclitaxel have been observed in patients whose tumors are resistant to 3-weekly paclitaxel. The level of activity of weekly paclitaxel for relapsed disease has led to its detailed evaluation in the first-line setting, and interest has been enhanced by the results of a Japanese Gynecological Oncology Group study that demonstrated a survival advantage for weekly paclitaxel compared with 3-weekly paclitaxel in combination with carboplatin as initial treatment. The enhanced efficacy of weekly paclitaxel may be due to greater drug exposure, a direct antiangiogenic effect, or both. Current research topics include the combination of weekly paclitaxel with molecular-targeted agents and the use of molecular profiling to better select patients for treatment.


Clinical Cancer Research | 2013

A Phase I Study of Quisinostat (JNJ-26481585), an Oral Hydroxamate Histone Deacetylase Inhibitor with Evidence of Target Modulation and Antitumor Activity, in Patients with Advanced Solid Tumors

B. Venugopal; Richard D. Baird; Rebecca Kristeleit; Ruth Plummer; R. Cowan; Adam Stewart; Nele Fourneau; Peter Hellemans; Yusri A. Elsayed; S. Mcclue; J.W. Smit; A. Forslund; C. Phelps; J. Camm; T. R. J. Evans; J. S. De Bono; Udai Banerji

Purpose: To determine the maximum-tolerated dose (MTD), dose-limiting toxicities (DLT), and pharmacokinetic and pharmacodynamic profile of quisinostat, a novel hydroxamate, pan-histone deacetylase inhibitor (HDACi). Experimental Design: In this first-in-human phase I study, quisinostat was administered orally, once daily in three weekly cycles to patients with advanced malignancies, using a two-stage accelerated titration design. Three intermittent schedules were subsequently explored: four days on/three days off; every Monday, Wednesday, Friday (MWF); and every Monday and Thursday (M-Th). Toxicity, pharmacokinetics, pharmacodynamics, and clinical efficacy were evaluated at each schedule. Results: Ninety-two patients were treated in continuous daily (2–12 mg) and three intermittent dosing schedules (6–19 mg). Treatment-emergent adverse events included: fatigue, nausea, decreased appetite, lethargy, and vomiting. DLTs observed were predominantly cardiovascular, including nonsustained ventricular tachycardia, ST/T-wave abnormalities, and other tachyarhythmias. Noncardiac DLTs were fatigue and abnormal liver function tests. The maximum plasma concentration (Cmax) and area under the plasma concentration–time curve (AUC) of quisinostat increased proportionally with dose. Pharmacodynamic evaluation showed increased acetylated histone 3 in hair follicles, skin and tumor biopsies, and in peripheral blood mononuclear cells as well as decreased Ki67 in skin and tumor biopsies. A partial response lasting five months was seen in one patient with melanoma. Stable disease was seen in eight patients (duration 4–10.5 months). Conclusions: The adverse event profile of quisinostat was comparable with that of other HDACi. Intermittent schedules were better tolerated than continuous schedules. On the basis of tolerability, pharmacokinetic predictions, and pharmacodynamic effects, the recommended dose for phase II studies is 12 mg on the MWF schedule. Clin Cancer Res; 19(15); 4262–72. ©2013 AACR.


British Journal of Haematology | 2002

Spontaneous remission of low-grade B-cell non-Hodgkin's lymphoma following withdrawal of methotrexate in a patient with rheumatoid arthritis: case report and review of the literature

Richard D. Baird; Richard van Zyl-Smit; Timothy Dilke; Sara E. Scott; Saad M. B. Rassam

Summary. A 69‐year‐old woman, who had suffered from deforming rheumatoid arthritis since the age of 40 years, had been treated with methotrexate for 3 years. She presented with a 7 week history of neck lymphadenopathy. Biopsy revealed low‐grade marginal‐zone B‐cell non‐Hodgkins lymphoma. Computerized tomography and bone marrow biopsy confirmed stage IIIA disease. Spontaneous complete remission of the lymphoma was achieved 14 months after withdrawing immune suppression with methotrexate.


Nature Reviews Clinical Oncology | 2016

Clinical development of new drug-radiotherapy combinations

Ricky A. Sharma; Ruth Plummer; Jk Stock; Trisha Greenhalgh; O Ataman; S Kelly; R Clay; Richard Alexander Adams; Richard D. Baird; Lucinda Billingham; Brown; S Buckland; H Bulbeck; Anthony J. Chalmers; G Clack; Amy Cranston; L Damstrup; Roberta Ferraldeschi; Forster; J Golec; Rm Hagan; Emma Hall; Ar Hanauske; Kevin J. Harrington; T Haswell; M. Hawkins; Tim Illidge; Hazel Jones; Andrew S. Kennedy; F. McDonald

In countries with the best cancer outcomes, approximately 60% of patients receive radiotherapy as part of their treatment, which is one of the most cost-effective cancer treatments. Notably, around 40% of cancer cures include the use of radiotherapy, either as a single modality or combined with other treatments. Radiotherapy can provide enormous benefit to patients with cancer. In the past decade, significant technical advances, such as image-guided radiotherapy, intensity-modulated radiotherapy, stereotactic radiotherapy, and proton therapy enable higher doses of radiotherapy to be delivered to the tumour with significantly lower doses to normal surrounding tissues. However, apart from the combination of traditional cytotoxic chemotherapy with radiotherapy, little progress has been made in identifying and defining optimal targeted therapy and radiotherapy combinations to improve the efficacy of cancer treatment. The National Cancer Research Institute Clinical and Translational Radiotherapy Research Working Group (CTRad) formed a Joint Working Group with representatives from academia, industry, patient groups and regulatory bodies to address this lack of progress and to publish recommendations for future clinical research. Herein, we highlight the Working Groups consensus recommendations to increase the number of novel drugs being successfully registered in combination with radiotherapy to improve clinical outcomes for patients with cancer.

Collaboration


Dive into the Richard D. Baird's collaboration.

Top Co-Authors

Avatar

J. S. De Bono

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Stan B. Kaye

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Olmos

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Johann S. de Bono

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Paul Workman

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Timothy A. Yap

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amita Patnaik

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Anthony W. Tolcher

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge