Network


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

Hotspot


Dive into the research topics where Mark Rothmann is active.

Publication


Featured researches published by Mark Rothmann.


Clinical Cancer Research | 2012

U.S. Food and Drug Administration Approval Summary: Brentuximab Vedotin for the Treatment of Relapsed Hodgkin Lymphoma or Relapsed Systemic Anaplastic Large-Cell Lymphoma

R. Angelo de Claro; Karen M. McGinn; Virginia E. Kwitkowski; Julie Bullock; Aakanksha Khandelwal; Bahru A. Habtemariam; Yanli Ouyang; Haleh Saber; Kyung Y. Lee; Kallappa M. Koti; Mark Rothmann; Marjorie A. Shapiro; Franciso Borrego; Kathleen Clouse; Xiao Hong Chen; Janice Brown; Lara Akinsanya; Robert C. Kane; Edvardas Kaminskas; Ann T. Farrell; Richard Pazdur

The U.S. Food and Drug Administration (FDA) describes the accelerated approval of brentuximab vedotin for patients with relapsed Hodgkin lymphoma and relapsed systemic anaplastic large-cell lymphoma (sALCL). FDA analyzed the results of two single-arm trials, enrolling 102 patients with Hodgkin lymphoma and 58 patients with sALCL. Both trials had primary endpoints of objective response rate (ORR) and key secondary endpoints of response duration and complete response (CR) rate. For patients with Hodgkin lymphoma, ORR was 73% (95% CI, 65–83%); median response duration was 6.7 months, and CR was 32% (95% CI, 23–42%). For patients with sALCL, ORR was 86% (95% CI, 77–95%), median response duration was 12.6 months, and CR was 57% (95% CI, 44–70%). The most common adverse reactions were neutropenia, peripheral sensory neuropathy, fatigue, nausea, anemia, upper respiratory infection, diarrhea, pyrexia, rash, thrombocytopenia, cough, and vomiting. FDA granted accelerated approval of brentuximab vedotin for the treatment of patients with Hodgkin lymphoma after failure of autologous stem cell transplantation (ASCT) or after failure of at least two prior multiagent chemotherapy regimens in patients who are not ASCT candidates, and for the treatment of patients with sALCL after failure of at least one prior multiagent chemotherapy regimen. Clin Cancer Res; 18(21); 5845–9. ©2012 AACR.


Clinical Cancer Research | 2013

U.S. Food and Drug Administration Approval: Carfilzomib for the Treatment of Multiple Myeloma

Thomas M. Herndon; Albert Deisseroth; Edvardas Kaminskas; Robert C. Kane; Kallappa M. Koti; Mark Rothmann; Bahru A. Habtemariam; Julie Bullock; Jeffrey D Bray; Jessica H Hawes; Todd R. Palmby; Josephine M. Jee; William M. Adams; Houda Mahayni; Janice Brown; Angelica Dorantes; Rajeshwari Sridhara; Ann T. Farrell; Richard Pazdur

The U.S. Food and Drug Administration (FDA) review leading to accelerated approval of carfilzomib is described. A single-arm trial enrolled 266 patients with multiple myeloma refractory to the most recent therapy who had received prior treatment with bortezomib and an immunomodulatory agent (IMID). Patients received carfilzomib by intravenous infusion over 2 to 10 minutes at a dose of 20 mg/m2 on days 1, 2, 8, 9, 15, and 16 of the 28 days of cycle 1, and at a dose of 27 mg/m2 on the same schedule in cycle 2 and subsequent cycles. The primary efficacy endpoint was overall response rate (ORR) as determined by an independent review committee using International Myeloma Working Group Uniform Response Criteria. The safety of carfilzomib was evaluated in 526 patients with multiple myeloma treated with various dosing regimens. The ORR was 23%. The median duration of response was 7.8 months. The most common adverse reactions associated with carfilzomib infusion were fatigue, anemia, nausea, thrombocytopenia, dyspnea, diarrhea, and fever. The most common serious adverse events were pneumonia, acute renal failure, fever, and congestive heart failure. Infusion reactions to carfilzomib could be reduced by pretreatment with dexamethasone and intravenous fluids. On July 20, 2012, the FDA granted accelerated approval of carfilzomib for the treatment of patients with multiple myeloma who have received at least two prior therapies including bortezomib and an IMID and who have shown disease progression while on therapy or within 60 days of completion of the last therapy. Clin Cancer Res; 19(17); 4559–63. ©2013 AACR.


Clinical Cancer Research | 2010

U.S. Food and Drug Administration Approval: Ofatumumab for the Treatment of Patients with Chronic Lymphocytic Leukemia Refractory to Fludarabine and Alemtuzumab

Steven Lemery; Jenny Zhang Zhang; Mark Rothmann; Jun Yang; Justin C. Earp; Hong Zhao; Andrew McDougal; Anne M. Pilaro; Raymond Chiang; Joseph E. Gootenberg; Patricia Keegan; Richard Pazdur

Purpose: To describe the data and analyses that led to the U.S. Food and Drug Administration (FDA) approval of ofatumumab (Arzerra, GlaxoSmithKline) for the treatment of patients with chronic lymphocytic leukemia (CLL) refractory to fludarabine and alemtuzumab. Experimental Design: The FDA reviewed the results of a planned interim analysis of a single-arm trial, enrolling 154 patients with CLL refractory to fludarabine, and a supportive dose-finding, activity-estimating trial in 33 patients with CLL. Patients in the primary efficacy study received ofatumumab weekly for eight doses, then every 4 weeks for an additional four doses; patients in the supportive trial received four weekly doses. In the primary efficacy study, endpoints were objective response rate and response duration. Results: For regulatory purposes, the primary efficacy population consisted of 59 patients with CLL refractory to fludarabine and alemtuzumab. In this subgroup, the investigator-determined objective response rate was 42% [99% confidence interval (CI), 26–60], with a median duration of response of 6.5 months (95% CI, 5.8–8.3); all were partial responses. The most common adverse reactions in the primary efficacy study were neutropenia, pneumonia, pyrexia, cough, diarrhea, anemia, fatigue, dyspnea, rash, nausea, bronchitis, and upper respiratory tract infections. Infusion reactions occurred in 44% of patients with the first infusion (300 mg) and 29% with the second infusion (2,000 mg). The most common serious adverse reactions were infections, neutropenia, and pyrexia. Conclusions: On October 26, 2009, the FDA granted accelerated approval to ofatumumab for the treatment of patients with CLL refractory to fludarabine and alemtuzumab, on the basis of demonstration of durable tumor shrinkage. Clin Cancer Res; 16(17); 4331–8. ©2010 AACR.


Clinical Cancer Research | 2008

U.S. Food and Drug Administration Approval: Panitumumab for Epidermal Growth Factor Receptor–Expressing Metastatic Colorectal Carcinoma with Progression Following Fluoropyrimidine-, Oxaliplatin-, and Irinotecan-Containing Chemotherapy Regimens

Ruthann M. Giusti; Kaushikkumar Shastri; Anne M. Pilaro; Chana Fuchs; Ruth Cordoba-Rodriguez; Kallappa M. Koti; Mark Rothmann; Angela Yuxin Men; Hong Zhao; Monica Hughes; Patricia Keegan; Karen D. Weiss; Richard Pazdur

Purpose: To describe the Food and Drug Administration review and marketing approval considerations for panitumumab (Vectibix) for the third-line treatment of patients with epidermal growth factor receptor–expressing metastatic colorectal carcinoma. Experimental Design: Food and Drug Administration reviewed a single, open-label, multicenter trial in which 463 patients with epidermal growth factor receptor–expressing metastatic colorectal cancer who had progressed on or following treatment with a regimen containing a fluoropyrimidine, oxaliplatin, and irinotecan were randomized (1:1) to receive best supportive care (BSC) with or without panitumumab (6 mg/kg every other week) administered until disease progression or intolerable toxicity. Progression and response were confirmed by an independent review committee masked to treatment assignment. At progression, patients in the BSC-alone arm were eligible to receive panitumumab. Results: Although median progression-free survival (PFS) was similar in both treatment arms (∼8 weeks), the mean PFS was ∼50% longer among patients receiving panitumumab than among those receiving BSC alone (96 versus 60 days, respectively) and the objective response rate in patients receiving panitumumab was 8%. However, no difference in overall survival was shown between the two study arms. Conclusions: Panitumumab received accelerated approval based on improvement in PFS and an independently confirmed response rate of 8%, similar to that observed with other active agents at this advanced stage of disease. Confirmation of clinical benefit will be required for full approval.


Clinical Cancer Research | 2012

U.S. Food and Drug Administration Approval: Ruxolitinib for the Treatment of Patients with Intermediate and High-Risk Myelofibrosis

Albert B. Deisseroth; Edvardas Kaminskas; Joseph A. Grillo; Wei Chen; Haleh Saber; Hong L. Lu; Mark Rothmann; Satjit Brar; Jian Wang; Christine Garnett; Julie Bullock; Laurie B. Burke; Atiqur Rahman; Rajeshwari Sridhara; Ann T. Farrell; Richard Pazdur

On November 16, 2011, the U.S. Food and Drug Administration (FDA) granted full approval to ruxolitinib, (Jakafi; Incyte Corp.), an inhibitor of the Janus kinases 1 and 2, for the treatment of patients with intermediate- or high-risk myelofibrosis, including primary myelofibrosis, postpolycythemia vera myelofibrosis, and postessential thrombocythemia myelofibrosis. This approval was based on the results of 2 large randomized phase III trials that enrolled patients with intermediate-2 or high-risk myelofibrosis and compared ruxolitinib with placebo (study 1) or best available therapy (study 2). The primary efficacy endpoint was the proportion of patients who experienced a reduction in spleen volume of ≥35% at 24 weeks (study 1) or 48 weeks (study 2). The key secondary endpoint in study 1 was the proportion of patients who experienced a ≥50% improvement from baseline in myelofibrosis total symptom score at 24 weeks. The results of these studies showed that a greater proportion of patients treated with ruxolitinib experienced a ≥35% reduction in spleen volume as compared with those treated with placebo (42% vs. 1%, P < 0.0001) or best available therapy (29% vs. 0%, P < 0.0001). A greater proportion of patients in study 1 experienced a ≥50% reduction in the myelofibrosis total symptom score during treatment with ruxolitinib than with placebo (46% vs. 5%, P < 0.0001). Ruxolitinib treatment was associated with an increased incidence of grades III and IV anemia, thrombocytopenia, and neutropenia. This is the first drug approved for myelofibrosis. Clin Cancer Res; 18(12); 3212–7. ©2012 AACR.


Clinical Trials | 2011

Some essential considerations in the design and conduct of non-inferiority trials.

Thomas R. Fleming; Katherine Odem-Davis; Mark Rothmann; Yuan Li Shen

Background Suppose a standard therapy (Standard) has been established to provide a clinically important reduction in risk of irreversible morbidity or mortality. In that setting, the safety and efficacy of an experimental intervention likely would be assessed in a clinical trial providing a comparison with Standard rather than a placebo arm. Such a trial often is designed to assess whether the efficacy of the experimental intervention is not unacceptably worse than that of Standard, and is called a non-inferiority trial. Formally, the non-inferiority trial usually is designed to rule out a non-inferiority margin, defined as the minimum threshold for what would constitute an unacceptable loss of efficacy. Purpose Even though the literature has many important articles identifying various approaches to the design and conduct of non-inferiority trials, confusion remains especially regarding key considerations for selecting the non-inferiority margin. The purpose of this article is to provide improved clarity regarding these considerations. Methods We present scientific insights into many factors that should be addressed in the design and conduct of non-inferiority trials to enhance their integrity and reliability, and provide motivation for key considerations that guide the selection of non-inferiority margins. We also provide illustrations and insights from recent experiences. Results Two considerations are essential, and should be addressed in separate steps, in the formulation of the non-inferiority margin. First, the margin should be formulated using adjustments to account for bias or lack of reliability in the estimate of the effect of Standard in the non-inferiority trial setting. Second, the non-inferiority margin should be formulated to achieve preservation of an appropriate percentage of the effect of Standard. Limitations The considerations, in particular regarding the importance of preservation of effect, might not apply to settings where it would be ethical as well as clinically relevant to include both Standard and placebo arms in the trial for direct comparisons with the experimental intervention arm. Conclusions Non-inferiority trials with non-rigorous margins allow substantial risk for accepting inadequately effective experimental regimens, leading to the risk of erosion in quality of health care. The design and conduct of non-inferiority trials, including selection of non-inferiority margins, should account for many factors that can induce bias in the estimated effect of Standard in the non-inferiority trial and thus lead to bias in the estimated effect of the experimental treatment, for the need to ensure the experimental treatment preserves a clinically acceptable fraction of Standards effect, and for the particular vulnerability of the integrity of a non-inferiority trial to the irregularities in trial conduct. Due to the inherent uncertainties in non-inferiority trials, alternative designs should be pursued whenever possible.


Clinical Cancer Research | 2008

Continuing Reassessment of the Risks of Erythropoiesis-Stimulating Agents in Patients with Cancer

Vinni Juneja; Patricia Keegan; Joseph E. Gootenberg; Mark Rothmann; Yuan Li Shen; Kyung Y. Lee; Karen D. Weiss; Richard Pazdur

Purpose: Erythropoiesis-stimulating agents (ESA) are approved for the treatment of anemia in patients with nonmyeloid malignancies whose anemia is due to the effect of concomitantly administered chemotherapy. Since the 1993 approval of epoetin alfa in patients with cancer, the risk of thrombovascular events, decreased survival, and poorer tumor control have been increasingly recognized. The risks of ESAs in patients with cancer and the design of trials to assess these risks have been the topic of discussion at two Oncologic Drugs Advisory Committees in 2004 and 2007. Experimental Design: Evaluation of randomized clinical trials comparing use of ESAs to transfusion support alone in patients with active cancer. Results: Six studies (Breast Cancer Erythropoeitin Survival Trial, Evaluation of NeoRecormon on outcome in Head And Neck Cancer in Europe, Danish Head and Neck Cancer, Lymphoid Malignancy, CAN-20, and Anemia of Cancer) investigating ESAs in oncology patients showed decreased survival, decreased duration of locoregional tumor control, and/or increased risk of thrombovascular events. In these six studies, ESA dosing was targeted to achieve and maintain hemoglobin values in excess of current recommendations, and in three of the six studies, ESAs were administered to patients not receiving chemotherapy. Conclusions: ESAs increase the risk of thrombovascular events and result in decreased survival and poorer tumor control when administered to achieve hemoglobin levels of ≥12 g/dL in patients with nonmyeloid malignancies. No completed or ongoing randomized, controlled trial has addressed safety issues of ESAs in patients with chemotherapy-associated anemia using currently approved dosing regimens in an epidermal tumor type. Additional studies are needed to better characterize these risks.


Oncologist | 2011

U.S. Food and Drug Administration Approval: Rituximab in Combination with Fludarabine and Cyclophosphamide for the Treatment of Patients with Chronic Lymphocytic Leukemia

Sandra J. Casak; Steven Lemery; Yuan Li Shen; Mark Rothmann; Aakanksha Khandelwal; Hong Zhao; Gina Davis; Vaishali Jarral; Patricia Keegan; Richard Pazdur

PURPOSE To describe the clinical studies that led to the FDA approval of rituximab in combination with fludarabine and cyclophosphamide (FC) for the treatment of patients with chronic lymphocytic leukemia (CLL). MATERIALS AND METHODS The results of two multinational, randomized trials in CLL patients comparing rituximab combined with fludarabine and cyclophosphamide versus FC were reviewed. The primary endpoint of both studies was progression-free survival (PFS). RESULTS The addition of rituximab to FC decreased the risk of a PFS event by 44% in 817 previously untreated patients and by 24% in 552 previously treated patients. Median survival times could not be estimated. Exploratory analysis in patients older than 70 suggested that there was no benefit to patients when rituximab was added to FC. The safety profile observed in both trials was consistent with the known toxicity profile of rituximab, FC, or CLL. CONCLUSIONS On the basis of the demonstration of clinically meaningful prolongation of PFS, the FDA granted regular approval to rituximab in combination with FC for the treatment of patients with CLL. The magnitude of the treatment effect in patients 70 years and older is uncertain.


Statistics in Biopharmaceutical Research | 2015

Statistical Considerations on Subgroup Analysis in Clinical Trials

Mohamed Alosh; Kathleen Fritsch; Mohammad F. Huque; Kooros Mahjoob; Gene Pennello; Mark Rothmann; Estelle Russek-Cohen; Fraser Smith; Stephen Wilson; Lilly Q. Yue

The objective of subgroup analysis of a clinical trial is to investigate consistency or heterogeneity of the treatment effect across subgroups, defined based on background characteristics. As such, subgroup analysis plays an essential role in the interpretation of the clinical trial findings. Consistency of treatment effect across trial subgroups indicates that the average treatment effect is in general applicable regardless of the specific background characteristics. Substantial heterogeneity in treatment effect may be indicative that treatment benefit pertains only to a subset of the population. However, heterogeneity in the observed treatment effect across subgroups can arise due to chance as a result of partitioning the population into several subgroups. Furthermore, as it is known, clinical trials are generally not powered for detecting heterogeneity, thus statistical tests may miss detection of existing heterogeneity due to low power. In this article, we aim to: (i) outline the major issues underlying subgroup analysis in clinical trials and provide general statistical guidance for interpretation of its findings, (ii) provide statistical perspectives on the design and analysis of a clinical trial that aims for establishing efficacy in a targeted subgroup along with that of its overall population, and (iii) highlight some of the underlying assumptions and issues relevant to Bayesian subgroup analysis, subgroup considerations for noninferiority trials, personalized medicine, subgroup misclassification, and finally, subgroup analysis for safety assessment.


Journal of Biopharmaceutical Statistics | 2003

On Non-inferiority Analysis Based On Delta-method Confidence Intervals

Mark Rothmann; Hsiao-Hui Tsou

For many indications where there is an effective standard therapy, active controlled trials are generally conducted when it is unethical to use a placebo. The efficacy objective of most such trials is the demonstration that the experimental therapy has superior efficacy to the active-control. The efficacy objective of a non-inferiority trial may be to rule out that the experimental treatment loses some prespecified fraction of the active-control effect. The size of the active-control effect may be based on previous trials comparing this active-control with a placebo—for example, through a meta-analysis. Delta-method 95% confidence interval procedures are among the testing procedures that have been proposed to test a non-inferiority hypothesis that an experimental treatment retains more than some prespecified fraction of the active-control effect. For time-to-event endpoints using hazard ratios, we will examine the type I error probability of such testing procedures under the assumption that the current active-control effect has been correctly modeled. Conditions are discussed for when such testing procedures maintain a desired approximate type I error rate and when such testing procedures will not. Two applications (one in Cardiorenalogy and one in Oncology) are given—one maintains the desired approximate type I error probability and the other does not. The delta-method 95% confidence interval procedures will also be contrasted with Fieller 95% confidence intervals. Testing based on Fieller 95% confidence intervals will maintain a desired approximate type I error rate. #The views expressed in this article do not necessarily represent those of the U.S. Food and Drug Administration.

Collaboration


Dive into the Mark Rothmann's collaboration.

Top Co-Authors

Avatar

Richard Pazdur

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Gang Chen

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Kallappa M. Koti

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Patricia Keegan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yuan Li Shen

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Edvardas Kaminskas

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

George Y. H. Chi

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Hong Zhao

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Kyung Y. Lee

Veterans Health Administration

View shared research outputs
Top Co-Authors

Avatar

Martin H. Cohen

Food and Drug Administration

View shared research outputs
Researchain Logo
Decentralizing Knowledge