Network


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

Hotspot


Dive into the research topics where John Rinehart is active.

Publication


Featured researches published by John Rinehart.


Journal of Clinical Oncology | 2004

Multicenter Phase II Study of the Oral MEK Inhibitor, CI-1040, in Patients With Advanced Non-Small-Cell Lung, Breast, Colon, and Pancreatic Cancer

John Rinehart; Alex A. Adjei; Patricia M. LoRusso; David Waterhouse; J. Randolph Hecht; Ronald B. Natale; Oday Hamid; Mary Varterasian; Peggy Asbury; Eric P. Kaldjian; Stephen Gulyas; David Y. Mitchell; Roman Herrera; Judith S. Sebolt-Leopold; Mark B. Meyer

PURPOSE This multicenter, open-label, phase II study was undertaken to assess the antitumor activity and safety of the oral mitogen-activated extracellular signal regulated kinase kinase (MEK) inhibitor, CI-1040, in breast cancer, colon cancer, non-small-cell lung cancer (NSCLC), and pancreatic cancer. PATIENTS AND METHODS Patients with advanced colorectal, NSCLC, breast, or pancreatic cancer received oral CI-1040 continuously at 800 mg bid. All patients had measurable disease at baseline, a performance status of 2 or less, and adequate bone marrow, liver, and renal function. Expression of pERK, pAkt, and Ki-67 was assessed in archived tumor specimens by quantitative immunohistochemistry. RESULTS Sixty-seven patients with breast (n = 14), colon (n = 20), NSCLC (n = 18), and pancreatic (n = 15) cancer received a total of 194 courses of treatment (median, 2.0 courses; range, one to 14 courses). No complete or partial responses were observed. Stable disease (SD) lasting a median of 4.4 months (range, 4 to 18 months) was confirmed in eight patients (one breast, two colon, two pancreas, and three NSCLC patients). Treatment was well tolerated, with 81% of patients experiencing toxicities of grade 2 or less severity. Most common toxicities included diarrhea, nausea, asthenia, and rash. A mild association (P < .055) between baseline pERK expression in archived tumor specimens and SD was observed. CONCLUSION CI-1040 was generally well tolerated but demonstrated insufficient antitumor activity to warrant further development in the four tumors tested. PD 0325901, a second generation MEK inhibitor, has recently entered clinical development and, with significantly improved pharmacologic and pharmaceutical properties compared with CI-1040, it may better test the therapeutic potential of MEK inhibition in cancer.


Clinical Cancer Research | 2010

Phase I Pharmacokinetic and Pharmacodynamic Study of the Oral MAPK/ERK Kinase Inhibitor PD-0325901 in Patients with Advanced Cancers

Patricia LoRusso; Smitha S. Krishnamurthi; John Rinehart; Lisle Nabell; Lisa Malburg; Paul B. Chapman; Samuel E. DePrimo; Steven Bentivegna; Keith D. Wilner; Weiwei Tan; Alejandro D. Ricart

Purpose: To determine tolerability, pharmacokinetics, and pharmacodynamics of PD-0325901, a highly potent, selective, oral mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase 1/2 inhibitor in advanced cancer patients. Experimental Design: Sixty-six patients received PD-0325901 at doses from 1 mg once daily to 30 mg twice daily (BID). Cycles were 28 days; three administration schedules were evaluated. Pharmacokinetic parameters were assessed and tumor biopsies were done to evaluate pharmacodynamics. Results: Common adverse events were rash, diarrhea, fatigue, nausea, and visual disturbances including retinal vein occlusion (RVO; n = 3). Neurotoxicity was frequent in patients receiving ≥15 mg BID. The maximum tolerated dose, 15 mg BID continuously, was associated with late-onset RVO outside the dose-limiting toxicity window. An alternative dose and schedule, 10 mg BID 5 days on/2 days off, was therefore expanded; one RVO event occurred. Three of 48 evaluable patients with melanoma achieved confirmed partial responses; 10 had stable disease ≥4 months. PD-0325901 exposure was generally dose proportional. Doses ≥2 mg BID consistently caused ≥60% suppression of phosphorylated ERK in melanoma. Fifteen patients showed significant decreases (≥50%) in Ki-67. Conclusions: PD-0325901 showed preliminary clinical activity. The maximum tolerated dose, based on first cycle dose-limiting toxicities, was 15 mg BID continuously. However, 10 and 15 mg BID continuous dosing and 10 mg BID 5 days on/2 days off schedules were associated with delayed development of RVO; thus, further enrollment to this trial was stopped. Intermittent dose scheduling between 2 and 10 mg BID should be explored to identify a recommended dose with long-term PD-0325901 use. Clin Cancer Res; 16(6); 1924–37


Clinical Cancer Research | 2004

Pretreatment with Dexamethasone Increases Antitumor Activity of Carboplatin and Gemcitabine in Mice Bearing Human Cancer Xenografts In Vivo Activity, Pharmacokinetics, and Clinical Implications for Cancer Chemotherapy

Hui Wang; Mao Li; John Rinehart; Ruiwen Zhang

Purpose: The present study was undertaken to determine the effects of dexamethasone (DEX) pretreatment on antitumor activity and pharmacokinetics of the cancer chemotherapeutic agents carboplatin and gemcitabine. Experimental Design: Antitumor activities of carboplatin and gemcitabine with or without DEX pretreatment were determined in six murine-human cancer xenograft models, including cancers of colon (LS174T), lung (A549 and H1299), and breast (MCF-7 and MDA-MB-468) and glioma (U87-MG). Effects of DEX on plasma and tissue pharmacokinetics of carboplatin and gemcitabine were also determined by using the LS174T, A549, and H1299 models. Results: Although DEX alone showed minimal antitumor activity, DEX pretreatment significantly increased the efficacy of carboplatin, gemcitabine, or a combination of both drugs by 2–4-fold in all xenograft models tested. Without DEX treatment, the tumor exposure to carboplatin, measured by the area under the curve, was markedly lower than normal tissues. However, DEX pretreatment significantly increased tumor carboplatin levels, including 200% increase in area under the curve, 100% increase in maximum concentration, and 160% decrease in clearance. DEX pretreatment similarly increased gemcitabine uptake in tumors. Conclusions: To our knowledge, this is the first report that DEX significantly enhances the antitumor activity of carboplatin and gemcitabine and increases their accumulation in tumors. These results provide a basis for further evaluation of DEX as a chemosensitizer in patients.


Journal of Clinical Oncology | 2002

Characterization of a Novel Prostate-Specific Antigen–Activated Peptide-Doxorubicin Conjugate in Patients With Prostate Cancer

Robert S. DiPaola; John Rinehart; John Nemunaitis; Scott Ebbinghaus; Eric J. Rubin; Terry Capanna; Marie Ciardella; S. Doyle-Lindrud; Susan Goodwin; Michelle Fontaine; Ned Adams; Adrienne Williams; Michael Schwartz; Greg Winchell; Karen E. Wickersham; Paul J. Deutsch; Siu-Long Yao

PURPOSE To evaluate safety and pharmacokinetics (PK), and determine the recommended dose for efficacy studies, of L-377202, a novel peptide conjugate of doxorubicin (Dox) that releases the active metabolites leucine-doxorubicin (Leu-Dox) and Dox on cleavage by membrane-bound prostate-specific antigen (PSA). PATIENTS AND METHODS Nineteen patients with advanced hormone-refractory prostate cancer were treated intravenously with 71 cycles of L-377202 at escalating dose levels of 20 (n = 1), 40 (n = 3), 80 (n = 4), 160 (n = 3), 225 (n = 6), and 315 mg/m(2) (n = 2) once every 3 weeks. Toxicity, response, and PK of L-377202 were assessed. RESULTS L-377202 was well tolerated. Dose-limiting grade 4 neutropenia was noted in two of two patients administered 315 mg/m(2) (both patients were able to resume therapy at 225 mg/m(2)). The recommended dose for efficacy studies was 225 mg/m(2), which induced grade 4 neutropenia in one of six patients. PK studies demonstrated that L-377202 was metabolized to Leu-Dox and Dox. PK were linear; after administration of single doses of 225 mg/m(2), the mean area under the concentration-time profiles of L-377202, Leu-Dox, and Dox were 6 micromol x L/h, 4 micromol x L/h, and 1 micromol x L/h, and peak concentrations were 14 micromol/L, 5 micromol/L, and 120 nmol/L, respectively. At 225 and 315 mg/m(2), five patients completed at least three cycles of therapy; two patients had a greater than 75% decrease in PSA, and one patient had a stabilized PSA. No response was noted at dose levels less than 225 mg/m(2). CONCLUSION This is the first study of selective drug delivery in humans using a novel PSA-activated agent. L-377202 was cleaved to produce detectable levels of the active metabolites Leu-Dox and Dox. L-377202 was well tolerated and established a safe dose level for further study.


Journal of Clinical Oncology | 1990

Phase II trial of interferon-beta-serine in metastatic renal cell carcinoma

Patricia Kinney; Pierre L. Triozzi; Donn C. Young; Joseph R. Drago; Brent C. Behrens; Henry A. Wise; John Rinehart

Interferon-beta-serine (IFN-beta-ser) is a muteine, recombinant IFN that is tolerated at a dose fivefold to 10-fold higher than IFN-alfa and interacts with the same cell membrane receptor as IFN-alfa. We hypothesized that at high doses IFN-beta-ser might induce a higher response rate than IFN-alfa in metastatic renal cell carcinoma. We undertook a phase II trial of IFN-beta-ser in patients with metastatic renal cell carcinoma. Patients were treated three times each week by a 2-hour intravenous infusion. Doses were escalated weekly (.25 to 5.5 mg, 1 mg = 180,000,000 U) until the maximum-tolerated treatment dose (MTTD) was determined. The MTTD is defined as one dose level less than that which caused grade 3 toxicity and was subsequently administered three times weekly for at least 4 weeks. Twenty-nine patients were entered, and 25 were assessable for response and toxicity. The performance status was 0-1 in all patients and only one patient received previous chemotherapy. The MTTD dose was 2.5 mg (range, 0.5 to 5.5 mg per treatment), although in 10 patients, doses were later deescalated because of cumulative toxicity. Initial dose-limiting toxicity and cumulative toxicity were fatigue, malaise, and fever in most patients. Hepatic transaminitis, neutropenia, and elevation of serum creatinine were also observed but were not dose-limiting. There was one complete response (CR) and four partial responses (PRs). All responses but one occurred in pulmonary metastases. The median time to response was 26 days (range, 17 to 102 days). These data demonstrate that IFN-beta-ser given in high doses exhibits significant antitumor activity in renal cell carcinoma; however, the objective response rate is 20%. This is no higher than previous IFN studies; therefore, we reject the hypothesis than IFN-beta-ser at high doses may induce a greater response rate than IFN-alfa. However, we did observe more responses than were seen in a similar trial undertaken with lower dose IFN-beta serine in renal cell carcinoma.


American Journal of Clinical Oncology | 2003

Hematopoietic protection by dexamethasone or granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients treated with carboplatin and ifosfamide.

John Rinehart; Lisa Keville; Jeff Neidhart; Lucas Wong; Lucio DiNunno; Patricia Kinney; Marie Aberle; Laura Tadlock; Gretchen A. Cloud

Based on preclinical studies, the authors undertook a pilot study to determine the hematologic and biologic effects of pretreatment with dexamethasone (Dex) or granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients receiving carboplatin and ifosfamide. Patients (n = 28) with metastatic solid tumors were randomized to receive pretreatment with Dex or GM-CSF or no pretreatment prior to courses 1 or 2 of carboplatin and ifosfamide. No alteration in dose of chemotherapy was allowed between course 1 and 2. Alterations of hematologic and nonhematologic toxicity and selected biologic parameters were compared between courses 1 and 2. Patients without any pretreatment demonstrated worsening hematologic toxicity in course 2 compared to course 1. In contrast, Dex pretreatment reduced hematopoietic toxicity and improved the absolute granulocyte count (AGC) and platelet count recovery times. For example, course 1 versus course 2 (with Dex pretreatment): AGC nadir (mm3) 153 versus 549 (p = 0.07), days AGC <500/mm3 7.8 versus 4.0 (p = 0.10), days to AGC recovery >1,500/mm3, 26 versus 22 (p = 0.034). Overall comparison between all five cohorts by analyses of variance demonstrated that intervention with Dex improved multiple hematopoietic toxicities, including AGC nadir (p = 0.015), and recovery times to AGC >1,500/mm3 (p = 0.07) and platelet count to >100,000/mm3 (p = 0.05). GM-CSF pretreatment did not worsen hematopoietic parameters after course 2 compared to course 1. Expected biologic effects of Dex and GM-CSF treatment were observed. Patients demonstrated an overall response rate of 32%, 1 complete response, and 8 partial responses. In patients with cancer, pretreatment with Dex or GM-CSF may significantly decrease the hematopoietic toxicity of chemotherapeutic agents.


Otolaryngology-Head and Neck Surgery | 2005

Neoadjuvant and concomitant chemotherapy and radiation therapy in patients with advanced head and neck carcinoma

John Rinehart; Tibor Ruff; Alan Cheung; Lewis Hutchinson; Roger Tuggle; D. Randall Pinkston; Lisa Keville; Lucas Wong

Our study evaluated the effectiveness of neoadjuvant chemotherapy and concomitant chemotherapy with radiotherapy compared to standard surgery and radiation therapy in patients with resectable stage III/IV head and neck squamous cell carcinoma. Forty-two eligible patients received neoadjuvant chemotherapy (cisplatin 100mg/m 2 intravenously day 1, and 5-fluorouracil 1g/m 2 /day continuous infusion days 1–5 every 3 weeks for 3 courses) followed by radiotherapy (65–70 Gy in 32–39 fractions to the primary site and lymph nodes; 50 Gy in 25–28 fractions to areas at risk) and concomitant chemotherapy (cisplatin 80mg/m 2 intravenously every 3 weeks starting on day 1 of radiotherapy). Neoadjuvant therapy induced grade 4 cytopenias (12/42 patients) and grade 4 gastrointestinal toxicities (7/42 patients). Concomitant radiochemotherapy-induced grade 4 toxicities (6 patients). Neoadjuvant chemotherapy biopsy-proven complete responses were 15 of 42 patients (36%), partial responses in 23 of 42 patients (55%), and an overall response rate of 91%. Thirty-seven of 38 responders received concomitant radiochemotherapy. Complete responses in 35 of 42 patients (83%), partial response in 7 of 42 patients (7%), and overall response in 90%. The 3-year disease-free and overall survival for chemotherapy plus radiotherapy versus surgery plus radiotherapy: 61% versus 43% (P = 0.17) and 71% versus 43% (P = 0.02). These data suggest that a randomized trial of concomitant radiochemotherapy versus neoadjuvant plus concomitant radiochemotherapy should be considered. EBM rating: B-2.


Brain Behavior and Immunity | 2005

80 Measurement of behavioral toxicity of carboplatin in mouse model

Elizabeth Kvale; Neil A. Accortt; Wayne A. Aldrich; John Rinehart; Pierre L. Triozzi

This study examined associations between cognitive functioning and immune measures (cytokine, complement) in chronic fatigue syndrome (CFS) patients. Methods. Eighteen CFS patients (mean length diagnosis = 73.1 months, SEM = 12.0) and 8 healthy controls completed measures of cognition (i.e., attention, sequencing, verbal and nonverbal learning, memory, and verbal fluency), complement activity, and cytokine activity (CFS subjects only). Results. Using demographically corrected t scores, the CFS patients performed worse than controls on Trailmaking Test B, Digit Symbol and the Stroop Color Word Test. Complement activity measured by C5a was significantly higher in CFS participants compared to controls, however, no differences were found between the groups on C3a, C4a or ECP. Cytokine data were not collected in control subjects for comparison. Several significant correlations between cognitive performance and immune measures were found for CFS participants. Increased IL-1b was related to higher scores on BVMT learning (r = .86, p = .025) and decline on Letter Fluency (r = .84, p = .036). Higher levels of IL-6 were related to better performance on Trailmaking Test A (r = .56, p = .049) and Trailmaking Test B (r = .58, p = .034), and higher levels of IFN-a were related to better performance on BVMT Recall (r = .56, p = .046). In CFS patients only, none of the complement activity was associated with cognitive test scores. In control subjects only, increased C3a was associated with lower Animal Fluency (r = .72, p = .043) lower BVMT Delay (r = .077, p = .025) and improved Trailmaking Test A (r = .816, p = .013). Higher C5a was associated with better Animal Fluency (r = .71, p = .05). Higher ECP was associated with lower Letter Fluency (r = .756, p = .03) and better BVMT learning (r = .84, p = .008). Conclusions. In CFS participants, increased IL-1b was related to lower verbal fluency. In contrast, better performance in visuomotor speed, visuomotor sequencing, and visual learning were related to higher levels of IL-1b, IL6 and IFN-a. These results suggest both positive and negative relationships between immune and cognitive functions in CFS patients. There were no significant relationships within the CFS group between cognition and complement activity. However, in control subjects, associations between complement activity and verbal fluency and verbal and nonverbal learning/memory scores suggest continued evaluation in this area is warranted. Overall, continued analysis with a larger sample size of CFS and control subjects may be useful in better understanding the mechanisms underlying the relationships between immune and cognitive functions.


Cancer Research | 1986

Phase I/II Trial of Human Recombinant β-Interferon Serine in Patients with Renal Cell Carcinoma

John Rinehart; Louis Malspeis; Donn C. Young; James A. Neidhart


Journal of the National Cancer Institute | 1991

Phase II Study of Intravenous Bolus Recombinant Interleukin-2 in Advanced Malignant Melanoma: Southwest Oncology Group Study

Robert P. Whitehead; Kenneth J. Kopecky; Michael K. Samson; John J. Costanzi; Ronald B. Natale; Lynn G. Feun; Evan M. Hersh; John Rinehart

Collaboration


Dive into the John Rinehart's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pierre L. Triozzi

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

James A. Neidhart

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gretchen A. Cloud

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Lisle Nabell

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patricia M. LoRusso

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge