Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Clin Cancer Res ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875108

ABSTRACT

On November 15, 2023, the U.S. Food and Drug Administration (FDA) granted traditional approval to repotrectinib (Augtyro®, Bristol Myers Squibb Corporation), for the treatment of adult patients with locally advanced or metastatic ROS1-positive non-small cell lung cancer (NSCLC). The approval was based on TRIDENT-1, a single arm trial with multiple cohorts of patients with ROS1 fusion-positive (hereafter "ROS1-positive") NSCLC, (NCT03093116), who were either treatment naïve or had received prior ROS1 TKI and/or platinum-based chemotherapy. The primary efficacy outcome measure is objective response rate (ORR) assessed by blinded independent central review (BICR) using response evaluation criteria in solid tumors (RECIST) version 1.1. ORR was assessed in 71 patients who were ROS1 TKI naïve and 56 patients who had received a prior ROS1 TKI. Among 71 patients who were ROS1 TKI naïve, the ORR was 79% (95% CI 68, 88); median duration of response was 34.1 months (95% CI 26, NE). In patients who had received a prior ROS1 TKI and no prior chemotherapy, the ORR was 38% (95% CI 25, 52). The median duration of response was 14.8 months (95% CI 7.6, NE) BICR-assessed responses were observed in CNS metastases in patients in both cohorts, and in patients who developed resistance mutations following prior TKI therapy. The most common (> 20%) adverse reactions were dizziness, dysgeusia, peripheral neuropathy, constipation, dyspnea, ataxia, fatigue, cognitive disorders, and muscular weakness. A unique feature of this ROS1 TKI approval is the inclusion of robust evidence of efficacy in patients with ROS1-positive NSCLC who had progressed on prior ROS1 TKIs.

2.
Regul Toxicol Pharmacol ; 149: 105616, 2024 May.
Article in English | MEDLINE | ID: mdl-38561147

ABSTRACT

Pharmacokinetic (PK) models are increasingly submitted to the FDA to support first-in-human (FIH) dose selection of immune-oncology products. To examine whether a simple PK modeling (SPM) using clearance for scaling was acceptable for dose estimation, FIH(SPM) doses were computed and compared to doses that were safely administered to patients. We concluded that the SPM approach is acceptable in FIH dose estimation, but the variables should be carefully selected for CD3 constructs. For CD3 constructs, use of 60 kg BWh, a clearance exponent of 0.75, and a targeted plasma concentration based on relevant and/or sensitive activity assays was an acceptable approach for FIH dose selection; use of 0.85 as the scaling factor is questionable at this time as it resulted in a FIH dose that was too close to the AHD for one product (7%). Immune activating mAbs were not sensitive to changes in the clearance exponent (0.75-0.85) or body weight (60-70 kg). For PD-1/PD-L1 mAbs, using products' in vitro EC50 in the model resulted in suboptimal FIH doses and clinical data of closely related products informed FIH dose selection. PK models submitted by sponsors were diverse in methods, assumptions, and variables, and the resulting FIH doses were not always optimal.


Subject(s)
Models, Biological , Humans , Dose-Response Relationship, Drug , B7-H1 Antigen/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/administration & dosage , Programmed Cell Death 1 Receptor/immunology , Neoplasms/immunology , Neoplasms/drug therapy , CD3 Complex/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/blood
3.
Lancet Oncol ; 24(10): e415-e423, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37797647

ABSTRACT

Anticancer agents can impair ovarian function, resulting in premature menopause and associated long-term health effects. Ovarian toxicity is not usually adequately assessed in trials of anticancer agents, leaving an important information gap for patients facing therapy choices. This American Society of Clinical Oncology (ASCO) statement provides information about the incorporation of ovarian toxicity measures in trial design. ASCO recommends: (1) measurement of ovarian toxicity in relevant clinical trials of anticancer agents that enrol post-pubertal, pre-menopausal patients; (2) collection of ovarian function measures at baseline and at 12-24 months after anticancer agent cessation, as a minimum, and later in line with the trial schedule; and (3) assessment of both clinical measures and biomarkers of ovarian function. ASCO recognises that routine measurement of ovarian toxicity and function in cancer clinical trials will add additional complexity and burden to trial resources but asserts that this issue is of such importance to patients that it cannot continue to be overlooked.


Subject(s)
Antineoplastic Agents , Neoplasms , Female , Humans , United States , Neoplasms/therapy , Antineoplastic Agents/adverse effects , Ovary , Medical Oncology
5.
Clin Cancer Res ; 28(19): 4173-4177, 2022 10 03.
Article in English | MEDLINE | ID: mdl-35679021

ABSTRACT

On September 17, 2021, the FDA approved cabozantinib (Cabometyx; Exelixis, Inc.) for the treatment of adult and pediatric patients 12 years of age and older with locally advanced or metastatic differentiated thyroid cancer (DTC) that has progressed following prior VEGFR-targeted therapy and who are radioactive iodine (RAI)-refractory or ineligible. This is the first approval for patients with RAI-refractory locally advanced or metastatic DTC who have progressed following prior therapy and the first approval in pediatric patients with DTC. The approval was based on data from COSMIC-311 (Study XL184-311, NCT03690388), an international, randomized, double-blind trial in which patients with locally advanced or metastatic RAI-refractory DTC that progressed during or following treatment with at least one VEGFR-targeting tyrosine kinase inhibitor were treated with either cabozantinib 60 mg orally once daily (N = 170) or placebo with best supportive care (N = 88). The primary efficacy outcome measures were progression-free survival (PFS) and overall response rate (ORR) by blinded independent central review per RECIST 1.1. The median PFS was 11.0 months [95% confidence interval (CI), 7.4-13.8] in the cabozantinib arm compared with 1.9 months (95% CI, 1.9-3.7) in the control arm, with an HR of 0.22 (95% CI, 0.15-0.31). The endpoint of ORR was not met. No new safety signals were identified with the exception of hypocalcemia, which was added as a warning in the product labeling.


Subject(s)
Adenocarcinoma , Anilides , Pyridines , Thyroid Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adult , Angiogenesis Inhibitors/therapeutic use , Anilides/adverse effects , Child , Humans , Iodine Radioisotopes/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/adverse effects , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology
6.
Clin Cancer Res ; 28(8): 1487-1492, 2022 04 14.
Article in English | MEDLINE | ID: mdl-34916216

ABSTRACT

On December 16, 2020, the FDA granted regular approval to margetuximab-cmkb (MARGENZA), in combination with chemotherapy, for the treatment of adult patients with HER2-positive (HER2+) metastatic breast cancer who have received two or more prior anti-HER2 regimens, at least one of which was for metastatic disease. Approval was based on data from SOPHIA, a multicenter, randomized, open-label, active controlled study comparing margetuximab with trastuzumab, in combination with chemotherapy. The primary efficacy endpoint was progression-free survival (PFS) by blinded independent central review. SOPHIA demonstrated a 0.9-month difference in median PFS between the two treatment arms [5.8 vs. 4.9 months, respectively; stratified HR, 0.76 (95% confidence interval: 0.59-0.98; P = 0.0334)]. Overall survival (OS) was immature at the data cut-off date of September 10, 2019. Infusion-related reactions (IRR) are an important safety signal associated with margetuximab plus chemotherapy. In SOPHIA, 13% of patients treated with margetuximab plus chemotherapy reported IRRs, of which 1.5% were grade 3. The most commonly reported adverse drug reactions (>10%) with margetuximab in combination with chemotherapy were fatigue/asthenia, nausea, diarrhea, vomiting, constipation, headache, pyrexia, alopecia, abdominal pain, peripheral neuropathy, arthralgia/myalgia, cough, decreased appetite, dyspnea, IRR, palmar-plantar erythrodysesthesia, and extremity pain. Overall, the favorable risk-benefit profile for margetuximab when added to chemotherapy supported its approval for the intended indication.


Subject(s)
Breast Neoplasms , Adult , Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Drug Approval , Female , Humans , Receptor, ErbB-2/therapeutic use , Trastuzumab/adverse effects
7.
Transgend Health ; 7(6): 514-520, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36644121

ABSTRACT

The prevalence of polycystic ovary syndrome (PCOS) among trans men has been reported as higher than among the cisgender population, which varies between 14.4% and 58%. In this cross-sectional study, we aimed to evaluate the association of oligo-anovulation and/or features of hyperandrogenism with the scores on the Utrecht Gender Dysphoria Scale (UGDS), the Body Image Scale (BIS), and the Short Form-36 Health Survey (SF-36) in treatment-naive trans men with PCOS seeking help for gender transition. The study sample consisted of trans men who were diagnosed with gender dysphoria (GD) between December 2019 and November 2020. To assess body dissatisfaction and psychological functioning, the UGDS, BIS, and SF-36 were administered to all transgender individuals as part of the routine assessment procedure. A total of 49 treatment-naive trans men were included in our study; 38.8% (n=19) of the participants were diagnosed with PCOS using the Rotterdam 2003 criteria. The scores of UGDS and BIS were significantly lower in the PCOS group compared with the non-PCOS group (p<0.001 and p=0.003, respectively). Among the eight parameters within SF-36, general health, emotional role, and vitality scores were significantly higher in the PCOS group (p=0.031, p=0.015, and p=0.006, respectively). Our study is the first study that demonstrates significantly lower GD, higher body image satisfaction and quality of life in trans men with PCOS compared with those without PCOS. Our findings are promising for larger prospective cohort studies to develop a consensus on the management of PCOS in trans men.

8.
Clin Cancer Res ; 27(9): 2378-2382, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33288660

ABSTRACT

On June 15, 2020, the FDA granted accelerated approval to lurbinectedin for the treatment of adult patients with metastatic small cell lung cancer (SCLC) with disease progression on or after platinum-based chemotherapy. Approval was granted on the basis of the clinically meaningful effects on overall response rate (ORR) and duration of response (DOR), and the safety profile observed in a multicenter, open-label, multicohort clinical trial (PM1183-B-005-14, NCT02454972), referred to as Study B-005, in patients with advanced solid tumors. The trial included a cohort of 105 patients with metastatic SCLC who had disease progression on or after platinum-based chemotherapy. The confirmed ORR determined by investigator assessment using RECIST 1.1 in the approved SCLC patient population was 35% [95% confidence interval (CI): 26-45], with a median DOR of 5.3 (95% CI: 4.1-6.4) months. The drug label includes warnings and precautions for myelosuppression, hepatotoxicity, and embryo-fetal toxicity. This is the first drug approved by the FDA in over 20 years in the second line for patients with metastatic SCLC. Importantly, this approval includes an indication for patients who have platinum-resistant disease, representing an area of particular unmet need.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbolines/therapeutic use , Drug Approval , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carbolines/pharmacology , Combined Modality Therapy , Disease Management , Drug Evaluation, Preclinical , Female , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Retreatment , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/mortality , Treatment Outcome , United States , United States Food and Drug Administration
9.
Support Care Cancer ; 28(11): 5455-5461, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32166381

ABSTRACT

PURPOSE: The use of mobile health (mHealth) technologies to augment patient care enables providers to communicate remotely with patients enhancing the quality of care and patient engagement. Few studies evaluated predictive factors of its acceptance and subsequent implementation, especially in medically underserved populations. METHODS: A cross-sectional study of 151 cancer patients was conducted at an academic medical center in the USA. A trained interviewer performed structured interviews regarding the barriers and facilitators of patients' current and desired use of mHealth technology for healthcare services. RESULTS: Of the 151 participants, 35.8% were male and ages ranged from 21 to 104 years. 73.5% of participants currently have daily access to internet, and 68.2% currently own a smartphone capable of displaying mobile applications. Among all participants, acceptability of a daily mHealth application was significantly higher in patients with a college-level degree (OR 2.78, CI95% 1.25-5.88) and lower in patients > 80 years of age (OR 0.05, CI95% 0.01-0.23). Differences in acceptability when adjusted for current smartphone use and daily access to internet were nonsignificant. Among smartphone users, the desire to increase cancer knowledge was associated with a higher likelihood of utilizing a mHealth application (OR 261.53, CI95% 10.13-6748.71). CONCLUSION: The study suggests that factors such as age, educational achievement, and access to internet are significant predictors of acceptability of a mHealth application among cancer patients. Healthcare organizations should consider these factors when launching patient engagement platforms.


Subject(s)
Internet/statistics & numerical data , Mobile Applications/statistics & numerical data , Neoplasms/psychology , Smartphone/statistics & numerical data , Telemedicine/statistics & numerical data , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Young Adult
10.
J Immunotoxicol ; 17(1): 67-85, 2020 12.
Article in English | MEDLINE | ID: mdl-32100588

ABSTRACT

Currently, there is a multitude of CD3 bispecifics with different molecular designs and binding properties in preclinical and clinical development for the treatment of liquid or solid tumors. The key safety concerns with CD3 bispecifics are excessive release of cytokines, which may translate to potentially life-threating cytokine release syndrome (CRS), target organ toxicity due to redirection of T-cells to normal tissues expressing the tumor-associated antigen (TAA) (off-tumor/on-target cytotoxicity), and, in some instances, neurotoxicity. Another key challenge is to arrive at a safe clinical starting dose and an efficient escalating strategy that allows patients in early dose cohorts the potential for clinical benefit in Phase 1 trials. To expand the therapeutic index and bring more treatment options to patients, there are intense efforts to overcome these challenges through improvements in molecular design, preclinical safety assessment strategies, and clinical management practices. A recent workshop at the U.S. Food and Drug Administration (FDA) with industry, academic, and regulatory agency representation was held to discuss the challenges and explore where such improvements to the development of CD3 bispecifics can be implemented. Here, the content of the presentations and the discussion that occurred during this workshop are summarized.


Subject(s)
Antibodies, Bispecific/toxicity , Antigens, Neoplasm/metabolism , Antineoplastic Agents/toxicity , CD3 Complex/antagonists & inhibitors , Cytokine Release Syndrome/prevention & control , Animals , Antibodies, Bispecific/administration & dosage , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , CD3 Complex/immunology , CD3 Complex/metabolism , Consensus , Consensus Development Conferences as Topic , Cytokine Release Syndrome/chemically induced , Cytokine Release Syndrome/immunology , Cytokines/metabolism , Drug Screening Assays, Antitumor/standards , Europe , Humans , Japan , Neoplasms/drug therapy , Neoplasms/immunology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Translational Research, Biomedical/standards , United States , United States Food and Drug Administration
11.
Regul Toxicol Pharmacol ; 110: 104511, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31678263

ABSTRACT

A systematic analysis of new commercial investigational new drug applications (IND) submitted to the FDA's Office of Hematology and Oncology Products (OHOP) in the Center for Drug Evaluation and Research was conducted to quantify the most common reasons INDs for oncology indications go on clinical hold. In OHOP, less than 10% of INDs went on hold or were withdrawn within the 30-day safety review period. Of INDs that were placed on hold, deficiencies were mainly clinical, followed by concerns related to pharmaceutical quality and nonclinical development. INDs were also characterized based on phase of development, product type, sponsors' regulatory experience, and occurrence of a pre-IND meeting. INDs that were placed on hold were mostly for first-in-human trials or submitted by sponsors with limited regulatory experience. INDs that went on hold or were safe-to-proceed had pre-IND meetings with comparable rates but sponsors with substantial experience appeared to benefit more from pre-IND meetings compared to those with limited experience. The time interval between the pre-IND meeting and the IND submission was longer for INDs that went on hold. To obtain useful FDA feedback on product development, it is essential to provide focused questions and supporting information in pre-IND meeting packages.


Subject(s)
Antineoplastic Agents , Investigational New Drug Application/statistics & numerical data , United States , United States Food and Drug Administration
12.
Regul Toxicol Pharmacol ; 107: 104429, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31325532

ABSTRACT

With a new generation of antibody-drug conjugates (ADCs) that contain a drug-to-antibody ratio (DAR) of 2, the question remains whether advances in technology have resulted in more stable and tumor-specific ADCs. These ADCs are anticipated to cause minimal systemic exposures of payloads, with toxicities being evident mainly at tumor sites. We examined 15 ADCs with PBD-dimer payloads and a DAR of 2 and concluded that dose limiting toxicities in animals and in humans are generally related to the payload. Both the payloads and the ADCs had pro-inflammatory responses causing severe toxicities that were at times of low incidence, making it difficult to assess a cause-effect relationship. Due to their low incidence, single-patient cohorts may not detect these events and such design may not be suitable in first-in-human (FIH) trials. The commonly proposed approach by the sponsors for FIH dose selection was 1/6th highest non-severely toxic dose (HNSTD) in monkeys. This approach resulted in an acceptable balance of safety and efficient dose escalation in phase 1 trials, when using data from repeat-dose toxicology studies and body surface area for scaling. No sponsor used the data generated in rodents or proposed novel approaches for FIH dose selection.


Subject(s)
Benzodiazepines/toxicity , Immunoconjugates/toxicity , Pyrroles/toxicity , Animals , Haplorhini , Humans , Maximum Tolerated Dose , Mice , Rats
13.
Int J Gynecol Cancer ; 28(8): 1478-1484, 2018 10.
Article in English | MEDLINE | ID: mdl-30045139

ABSTRACT

OBJECTIVE: Venous thromboembolism (VTE) is a major cause of mortality and morbidity in hospitalized patients with malignancy. Nationwide Inpatient Sample database was analyzed to determine the trends in the rate of hospitalization and mortality from VTE in hospitalized ovarian cancer patients and assess its economic impact and resource utilization. METHOD: We queried the 2003 to 2011 Nationwide Inpatient Sample database from Healthcare Cost and Utilization project (Agency of Healthcare Research and Quality) to identify all adults (age ≥18 years) ovarian cancer. Patients hospitalized with VTE as one of the top 3 discharge diagnoses were also identified. Demographic characteristics and in-hospital outcomes of this population were compared with ovarian cancer patients without VTE. Binary logistic regression analysis was used to obtain adjusted odds ratios (ORs). RESULTS: A total of 34,249 (3.5%) of a total of 981,386 hospitalized ovarian cancer patients had an accompanying diagnosis of VTE. Mean age of the study population was 64 years. After adjusting for potential confounders, compared with those without VTE, ovarian cancer patients with VTE had significantly higher inpatient mortality (6.2% vs 4.3%; OR, 1.12 [confidence interval (CI), 1.06-1.17]; P < .001), longer length of stay (5 vs 4 days; OR, 1.40 [CI, 1.36-1.43]; P < .001), higher average cost of hospitalization (US $26,000 vs US $22,000; OR, 1.10 [CI, 1.07-1.13]; P < .001), and greater disability at discharge (OR, 1.34 [CI, 1.31-1.38]; P < .001). Although the annual number of VTE admissions in ovarian cancer patients increased, in-hospital mortality declined from 10.9% in 2003 to 5.3% in 2011. CONCLUSIONS: Venous thromboembolism in hospitalized patients with ovarian cancer is associated with higher inpatient mortality, length of stay, higher cost of hospitalization, and disability at discharge. The hospitalization rate has increased, but the inpatient mortality rate has declined over study period.


Subject(s)
Ovarian Neoplasms/epidemiology , Venous Thromboembolism/epidemiology , Aged , Cohort Studies , Databases, Factual , Female , Hospital Mortality/trends , Hospitalization/economics , Hospitalization/trends , Humans , Length of Stay/trends , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/economics , Ovarian Neoplasms/mortality , Prognosis , United States/epidemiology , Venous Thromboembolism/economics , Venous Thromboembolism/mortality
14.
Case Rep Neurol Med ; 2018: 9281918, 2018.
Article in English | MEDLINE | ID: mdl-30652037

ABSTRACT

OBJECTIVES: The increased risk of thromboembolic complications with active cancer is well known. We present this case to highlight that chemotherapy may increase the risk of thromboembolic events even further in cancer patients. METHODS: We report a case of a 64-year-old male with Diffuse Large B-Cell Non-Hodgkin's Lymphoma who presented with left-sided headache and right calf pain two weeks after starting Rituximab/Gemcitabine/Cisplatin/Dexamethasone chemotherapy. Neurological examination was normal, but there was an absent right dorsalis pedis pulse. He subsequently developed left vision loss. CT angiogram of the head and neck revealed occlusion of his left internal carotid artery and poor opacification of the left ophthalmic artery. An angiogram of the right leg further revealed acute occlusion of the popliteal artery. RESULTS: The patient underwent intra-arterial Tissue Plasminogen Activator injection to his lower limb and was started on Low Molecular Weight Heparin. His vision gradually recovered with time. His chemotherapy regimen was changed to RICE (Rituximab, Ifosfamide, Carboplatin, Etoposide). CONCLUSION: Based on literature review, there are numerous similar presentations of arterial thromboembolism in patients on Cisplatin-based chemotherapy. A high index of suspicion for such events should be maintained for patients on chemotherapy presenting with unusual symptoms.

15.
Regul Toxicol Pharmacol ; 90: 144-152, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28887049

ABSTRACT

We retrospectively examined the nonclinical studies conducted with 17 CD3 bispecific constructs in support of first-in-human (FIH) trials in oncology. We also collected information on the design of dose-finding clinical trials. Sponsors have used different MABEL approaches for FIH dose selection. To better assess acceptable approaches, FIH doses were computed from nonclinical studies and compared to the maximum tolerated doses (MTDs) in patients, to the highest human doses (HHDs) when an MTD was not identified, or to the recommended human dose (RHD) for blinatumomab. We concluded that approaches based on receptor occupancy, highest non-severely toxic dose, or no-observed adverse effect level are not acceptable for selecting the FIH dose as they resulted in doses close to or above the MTDs, HHDs, or the RHD. A FIH dose corresponding to 10%-30% pharmacologic activity (PA) was an acceptable approach. A FIH dose corresponding to 50% PA was acceptable for all except one construct, potentially due to its biological or structural properties. The most common toxicities in animals and patients were those related to cytokine release. Doses were better tolerated when intra-animal or intra-patient dose escalation was used. Exposing naïve patients to an MTD achieved with intra-patient dose escalation design may be unsafe.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antineoplastic Agents/administration & dosage , CD3 Complex/antagonists & inhibitors , Maximum Tolerated Dose , No-Observed-Adverse-Effect Level , Animals , Antibodies, Bispecific/toxicity , Antineoplastic Agents/toxicity , Clinical Trials as Topic , Dose-Response Relationship, Drug , Humans , Primates , United States , United States Food and Drug Administration
16.
Regul Toxicol Pharmacol ; 81: 448-456, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27743776

ABSTRACT

As sub-therapeutic doses are not medically justifiable in patients with cancer, we retrospectively analyzed data on immune activating products, to assess approaches used in first-in-human (FIH) dose selection, the utility of animal toxicology studies in dose selection, and the length of time to complete FIH trials. The information collected included pharmacology and toxicology data, FIH dose and rationale, and dose-finding trial design. We used the principles of the Hill equation to estimate the FIH doses for antibodies and compared them to the doses administered to patients with acceptable toxicities. For approximately half the antibodies (44%) examined, the FIH doses were at least a hundred-fold lower than the doses safely administered to patients, indicating optimization of FIH dose selection and/or optimization of dose-finding trial design is needed to minimize patient exposure to sub-therapeutic doses. However, selection of the FIH dose for antibodies based on animal toxicology studies using 1/6th the HNSTD or 1/10th the NOAEL resulted in human doses that were unsafe for several antibodies examined. We also concluded that antibodies with Fc-modifications for increased effector function may be less tolerated, resulting in toxicities at lower doses than those without such modifications. There was insufficient information to evaluate CD3 bispecific products.


Subject(s)
Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Neoplasms/immunology , United States Food and Drug Administration , Animals , Antibodies, Monoclonal/administration & dosage , Dose-Response Relationship, Drug , Humans , United States
18.
Regul Toxicol Pharmacol ; 71(3): 444-52, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25661711

ABSTRACT

Antibody-drug conjugates (ADCs) are complex molecules composed of monoclonal antibodies conjugated to potent cytotoxic agents through chemical linkers. Because of this complexity, sponsors have used different approaches for the design of nonclinical studies to support the safety evaluation of ADCs and first-in-human (FIH) dose selection. We analyzed this data with the goal of describing the relationship between nonclinical study results and Phase 1 study outcomes. We summarized the following data from investigational new drug applications (INDs) for ADCs: plasma stability, animal study designs and toxicities, and algorithms used for FIH dose selection. Our review found that selecting a FIH dose that is 1/6th the highest non-severely toxic dose (HNSTD) in cynomolgus monkeys or 1/10th the STD10 in rodents scaled according to body surface area (BSA) generally resulted in the acceptable balance of safety and efficient dose-escalation in a Phase 1 trial. Other approaches may also be acceptable, e.g. 1/10th the HNSTD in monkeys using BSA or 1/10th the NOAEL in monkeys or rodents using body weight for scaling. While the animal data for the vc-MMAE platform yielded variable range of HNSTDs in cynomolgus monkeys, MTDs were in a narrow range in patients, suggesting that for ADCs sharing the same small molecule drug, linker and drug:antibody ratio, prior clinical data can inform the design of a Phase 1 clinical trial.


Subject(s)
Antineoplastic Agents/toxicity , Drug Approval , Immunoconjugates/toxicity , Toxicity Tests , United States Food and Drug Administration , Algorithms , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Body Surface Area , Body Weight , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Drug Dosage Calculations , Drug Stability , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/blood , Macaca fascicularis , Maximum Tolerated Dose , Mice , Models, Animal , No-Observed-Adverse-Effect Level , Rats , Reproducibility of Results , Risk Assessment , Toxicity Tests/methods , United States
19.
Curr Urol ; 7(4): 210-3, 2014 Oct.
Article in English | MEDLINE | ID: mdl-26195954

ABSTRACT

Non-Hodgkin's lymphoma (NHL) represents 4% of newly diagnosed cancer in 2013 with a 59-82% 5-year survival depending on the tumor location. Primary presentation of lymphoma consists of lymphadenopathy or swelling of the lymph nodes and non-specific systemic symptoms such as fevers, night sweats, and weight loss. Less commonly, NHL arises from non-lymphoid tissue. We report a unique case of NHL arising from the ureteral wall which was visualized via non-contrast CT and direct vision through ureteroscopy.

20.
Clin Cancer Res ; 19(22): 6067-73, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24141628

ABSTRACT

This article summarizes the regulatory evaluation that led to the full approval of enzalutamide (XTANDI, Medivation Inc.) by the U.S. Food and Drug Administration (FDA) on August 31, 2012, for the treatment of patients with metastatic castration-resistant prostate cancer who have previously received docetaxel. This approval was based on the results of a randomized, placebo-controlled trial which randomly allocated 1,199 patients with mCRPC who had received prior docetaxel to receive either enzalutamide, 160 mg orally once daily (n = 800), or placebo (n = 399). All patients were required to continue androgen deprivation therapy. The primary endpoint was overall survival. At the prespecified interim analysis, a statistically significant improvement in overall survival was demonstrated for the enzalutamide arm compared with the placebo arm [HR = 0.63; 95% confidence interval: 0.53-0.75; P < 0.0001]. The median overall survival durations were 18.4 months and 13.6 months in the enzalutamide and placebo arms, respectively. The most common adverse reactions (≥10%) included asthenia or fatigue, back pain, diarrhea, arthralgia, hot flush, peripheral edema, musculoskeletal pain, headache, and upper respiratory infection. Seizures occurred in 0.9% of patients on enzalutamide compared with no patients on the placebo arm. Overall, the FDA's review and analyses of the submitted data confirmed that enzalutamide had a favorable benefit-risk profile in the study patient population, thus supporting its use for the approved indication. The recommended dose is 160 mg of enzalutamide administered orally once daily. Enzalutamide represents the third product that the FDA has approved in the same disease setting within a period of 2 years. Clin Cancer Res; 19(22); 6067-73. ©2013 AACR.


Subject(s)
Drug Approval , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/mortality , Adult , Aged , Aged, 80 and over , Benzamides , Docetaxel , Humans , Male , Middle Aged , Nitriles , Phenylthiohydantoin/adverse effects , Phenylthiohydantoin/therapeutic use , Survival , Taxoids/therapeutic use , Treatment Outcome , United States , United States Food and Drug Administration
SELECTION OF CITATIONS
SEARCH DETAIL
...