Parathyroid carcinoma (PC) is a rare malignancy, often characterized by the unregulated secretion of parathyroid hormone. The sequelae of severe hypercalcemia together with direct complications from tumor dissemination in patients with advanced disease are usually fatal. Due to its rarity, formal studies to guide the diagnosis and management of parathyroid carcinoma are lacking. However, recent data from case reports, case series, and registry studies suggest the emergence of new and effective treatment approaches for this understudied disease. We reviewed existing literature on the diagnosis and management of parathyroid carcinoma. Our findings suggest that traditional approaches such as surgical resection for both localized and metastatic diseases continue to play an important role in patient management. For patients with unresectable disease, newer systemic treatment approaches, including the use of temozolomide and tyrosine kinase inhibitors, may offer clinical benefit.
{"title":"Diagnosis and Management of Parathyroid Carcinoma.","authors":"Alexander Lazzaro, Grace Qing Zhao, Matthew Kulke","doi":"10.1002/cpt.3432","DOIUrl":"https://doi.org/10.1002/cpt.3432","url":null,"abstract":"<p><p>Parathyroid carcinoma (PC) is a rare malignancy, often characterized by the unregulated secretion of parathyroid hormone. The sequelae of severe hypercalcemia together with direct complications from tumor dissemination in patients with advanced disease are usually fatal. Due to its rarity, formal studies to guide the diagnosis and management of parathyroid carcinoma are lacking. However, recent data from case reports, case series, and registry studies suggest the emergence of new and effective treatment approaches for this understudied disease. We reviewed existing literature on the diagnosis and management of parathyroid carcinoma. Our findings suggest that traditional approaches such as surgical resection for both localized and metastatic diseases continue to play an important role in patient management. For patients with unresectable disease, newer systemic treatment approaches, including the use of temozolomide and tyrosine kinase inhibitors, may offer clinical benefit.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131307","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sharif I Soleman, Juan Maya, Paul Levesque, Atif Mohammad, Lisa Christopher, Justin Schumacher, Aparna Nanduri, Pitchumani Sivakumar, Marc Kozinn, Philippe Costet, Chang Wang, Jeremy Richter, Dara Hawthorne, Anh Bui, Veena S Rao, Daniel Dickerson, Jeffrey Testani, Francisco Ramírez-Valle, Frédéric Baribaud, Bindu Murthy, Samira Merali
In patients with heart failure (HF) who respond inadequately to loop diuretic therapy, BMS-986308, an oral, selective, reversible renal outer medullary potassium channel (ROMK) inhibitor may represent an effective diuretic with a novel mechanism of action. We present data from the first-in-human study aimed to assess the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) following single ascending doses of BMS-986308 in healthy adult participants. Forty healthy participants, aged from 20 to 55 years, and body mass index (BMI) from 19.8 to 31.6 kg/m2 were assigned to 1 of 5 dose cohorts (1, 3, 10, 30, and 100 mg) and randomized (6:2) to receive BMS-986308 oral solution or matching placebo. Following administration, BMS-986308 was rapidly absorbed with a median time to maximum concentration (Tmax) of 1.00 to 1.75 h and exhibiting a mean terminal half-life (t1/2) of approximately 13 h. Dose proportionality was evident in BMS-986308 area under the concentration-time curve (AUC), while maximum concentration (Cmax) was slightly greater than dose-proportional. We observed that urine output (or diuresis; mL) and urinary sodium excretion (or natriuresis; mmol) increased in a dose-dependent manner, starting at a minimum pharmacologically active dose of 30 mg. The largest mean changes from baseline in diuresis and natriuresis occurred in both the 6- and -24 h post-dose period following administration of 100 mg (1683.0 mL and 2055.3 mL, and 231.7 mmol and 213.7 mmol, respectively; ***P < 0.001). Overall, single-dose BMS-986308 was found to be safe, well-tolerated, with an excellent PK profile, and substantial diuretic and natriuretic activity.
{"title":"First-in-Human Study to Assess the Safety, Pharmacokinetics, and Pharmacodynamics of BMS-986308: A Renal Outer Medullary Potassium Channel Inhibitor.","authors":"Sharif I Soleman, Juan Maya, Paul Levesque, Atif Mohammad, Lisa Christopher, Justin Schumacher, Aparna Nanduri, Pitchumani Sivakumar, Marc Kozinn, Philippe Costet, Chang Wang, Jeremy Richter, Dara Hawthorne, Anh Bui, Veena S Rao, Daniel Dickerson, Jeffrey Testani, Francisco Ramírez-Valle, Frédéric Baribaud, Bindu Murthy, Samira Merali","doi":"10.1002/cpt.3430","DOIUrl":"https://doi.org/10.1002/cpt.3430","url":null,"abstract":"<p><p>In patients with heart failure (HF) who respond inadequately to loop diuretic therapy, BMS-986308, an oral, selective, reversible renal outer medullary potassium channel (ROMK) inhibitor may represent an effective diuretic with a novel mechanism of action. We present data from the first-in-human study aimed to assess the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) following single ascending doses of BMS-986308 in healthy adult participants. Forty healthy participants, aged from 20 to 55 years, and body mass index (BMI) from 19.8 to 31.6 kg/m<sup>2</sup> were assigned to 1 of 5 dose cohorts (1, 3, 10, 30, and 100 mg) and randomized (6:2) to receive BMS-986308 oral solution or matching placebo. Following administration, BMS-986308 was rapidly absorbed with a median time to maximum concentration (T<sub>max</sub>) of 1.00 to 1.75 h and exhibiting a mean terminal half-life (t<sub>1/2</sub>) of approximately 13 h. Dose proportionality was evident in BMS-986308 area under the concentration-time curve (AUC), while maximum concentration (C<sub>max</sub>) was slightly greater than dose-proportional. We observed that urine output (or diuresis; mL) and urinary sodium excretion (or natriuresis; mmol) increased in a dose-dependent manner, starting at a minimum pharmacologically active dose of 30 mg. The largest mean changes from baseline in diuresis and natriuresis occurred in both the 6- and -24 h post-dose period following administration of 100 mg (1683.0 mL and 2055.3 mL, and 231.7 mmol and 213.7 mmol, respectively; ***P < 0.001). Overall, single-dose BMS-986308 was found to be safe, well-tolerated, with an excellent PK profile, and substantial diuretic and natriuretic activity.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142102605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jae Hyeon Yu, Sangwon Lee, Yoon Jung Kim, Won Young Kim, Min Jung Lee, Yun Kim
The U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) oversee pharmaceutical regulations, including orphan drugs targeting rare diseases with limited patient populations. Post-marketing studies are crucial for monitoring safety and efficacy, with post-marketing requirements (PMRs) mandated by the regulatory agencies to ensure compliance. This study aims to compare PMR statuses, objectives, and pivotal trial characteristics of orphan drugs approved by the FDA (n = 154) and EMA (n = 79) from 2008 to 2018, shedding light on regulatory differences and their impact on drug development. Contrary to expectations, our analysis found no significant disparity in the proportion of orphan drugs with and without PMRs approved by both the FDA (48.1%) and EMA (55.7%). Safety concerns surrounding orphan drugs post-approval, attributed partly to pivotal trial design, underscore the need for robust post-marketing surveillance. While the FDA primarily focuses on post-marketing safety (36.1%), the EMA places a higher emphasis on both efficacy and safety (47.1%), reflecting distinct approaches to PMR management between the two regulatory bodies. The observed trend of delayed PMRs at the EMA (47.1%) highlights the importance of effective cooperation between regulators and pharmaceutical companies to ensure the timely completion of PMRs and enhance drug safety.
{"title":"Assessing Post-Marketing Requirements for Orphan Drugs: A Cross-Sectional Analysis of FDA and EMA Oversight.","authors":"Jae Hyeon Yu, Sangwon Lee, Yoon Jung Kim, Won Young Kim, Min Jung Lee, Yun Kim","doi":"10.1002/cpt.3397","DOIUrl":"https://doi.org/10.1002/cpt.3397","url":null,"abstract":"<p><p>The U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) oversee pharmaceutical regulations, including orphan drugs targeting rare diseases with limited patient populations. Post-marketing studies are crucial for monitoring safety and efficacy, with post-marketing requirements (PMRs) mandated by the regulatory agencies to ensure compliance. This study aims to compare PMR statuses, objectives, and pivotal trial characteristics of orphan drugs approved by the FDA (n = 154) and EMA (n = 79) from 2008 to 2018, shedding light on regulatory differences and their impact on drug development. Contrary to expectations, our analysis found no significant disparity in the proportion of orphan drugs with and without PMRs approved by both the FDA (48.1%) and EMA (55.7%). Safety concerns surrounding orphan drugs post-approval, attributed partly to pivotal trial design, underscore the need for robust post-marketing surveillance. While the FDA primarily focuses on post-marketing safety (36.1%), the EMA places a higher emphasis on both efficacy and safety (47.1%), reflecting distinct approaches to PMR management between the two regulatory bodies. The observed trend of delayed PMRs at the EMA (47.1%) highlights the importance of effective cooperation between regulators and pharmaceutical companies to ensure the timely completion of PMRs and enhance drug safety.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142078625","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Stef Schouwenburg, Fleur M Keij, Gerdien A Tramper-Stranders, René F Kornelisse, Irwin K M Reiss, Pieter A J G de Cock, Evelyn Dhont, Kevin M Watt, Anouk E Muller, Robert B Flint, Birgit C P Koch, Karel Allegaert, Tim Preijers
Data published on the oral clavulanic acid pharmacokinetics in the pediatric population is lacking. This research aimed to describe clavulanic acid disposition following oral and intravenous administration and to provide insights into clavulanic acid exposure based on threshold concentrations for (pre-)term neonates and infants. This pooled population pharmacokinetic study combined four datasets for analysis in NONMEM v7.4.3. Clavulanic acid exposure was simulated using the percentage of time above the threshold concentrations (%fT > CT). Multiple dosage regimens and amoxicillin/clavulanic acid dosage ratios were evaluated. The cohort consisted of 89 (42 oral, 47 intravenous) subjects (403 samples) with a median (range) postnatal age 54.5 days (0-365), gestational age 37.4 weeks (23.0-41.7), and current bodyweight 3.9 kg (0.6-9.0). A one-compartment model with first-order absorption best described clavulanic acid pharmacokinetics with postnatal age as a covariate on the inter-individual variability of clearance. Oral bioavailability was 24.4% in neonates up to 10 days of age. An oral dosing regimen 90 mg/kg/day amoxicillin/clavulanic acid (4:1 ratio) resulted in 40.2% of simulated patients achieving 100% fT > CT,2mg/L. An amoxicillin/clavulanic acid ratio of 4:1 is preferred for neonatal oral regimens due to the higher exposure along the entire %fT > CT range (0-100%) as ratios higher than 4:1 might result in inadequate exposure. Our results highlight substantial exposure differences (%fT > CT) when using threshold concentrations of 1 mg/L vs. 2 mg/L. This first population pharmacokinetic model for clavulanic acid in neonates may serve as a foundational step for future research, once more precise clavulanic acid targets become available.
{"title":"A Pooled Population Pharmacokinetic Study of Oral and Intravenous Administration of Clavulanic Acid in Neonates and Infants: Targeting Effective Beta-Lactamase Inhibition.","authors":"Stef Schouwenburg, Fleur M Keij, Gerdien A Tramper-Stranders, René F Kornelisse, Irwin K M Reiss, Pieter A J G de Cock, Evelyn Dhont, Kevin M Watt, Anouk E Muller, Robert B Flint, Birgit C P Koch, Karel Allegaert, Tim Preijers","doi":"10.1002/cpt.3423","DOIUrl":"https://doi.org/10.1002/cpt.3423","url":null,"abstract":"<p><p>Data published on the oral clavulanic acid pharmacokinetics in the pediatric population is lacking. This research aimed to describe clavulanic acid disposition following oral and intravenous administration and to provide insights into clavulanic acid exposure based on threshold concentrations for (pre-)term neonates and infants. This pooled population pharmacokinetic study combined four datasets for analysis in NONMEM v7.4.3. Clavulanic acid exposure was simulated using the percentage of time above the threshold concentrations (%fT > C<sub>T</sub>). Multiple dosage regimens and amoxicillin/clavulanic acid dosage ratios were evaluated. The cohort consisted of 89 (42 oral, 47 intravenous) subjects (403 samples) with a median (range) postnatal age 54.5 days (0-365), gestational age 37.4 weeks (23.0-41.7), and current bodyweight 3.9 kg (0.6-9.0). A one-compartment model with first-order absorption best described clavulanic acid pharmacokinetics with postnatal age as a covariate on the inter-individual variability of clearance. Oral bioavailability was 24.4% in neonates up to 10 days of age. An oral dosing regimen 90 mg/kg/day amoxicillin/clavulanic acid (4:1 ratio) resulted in 40.2% of simulated patients achieving 100% fT > C<sub>T,2mg/L</sub>. An amoxicillin/clavulanic acid ratio of 4:1 is preferred for neonatal oral regimens due to the higher exposure along the entire %fT > C<sub>T</sub> range (0-100%) as ratios higher than 4:1 might result in inadequate exposure. Our results highlight substantial exposure differences (%fT > C<sub>T</sub>) when using threshold concentrations of 1 mg/L vs. 2 mg/L. This first population pharmacokinetic model for clavulanic acid in neonates may serve as a foundational step for future research, once more precise clavulanic acid targets become available.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142102604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cécile Ollivier, Solange Corriol-Rohou, Marta Del Álamo, Roseline Favresse, Johanna Kostenzer, Mathieu Boudes, Anton E Ussi, Klaus Viel, R Michael Linden, Magda Chlebus
{"title":"The Rare Disease Moonshot: Paradigms Shift, Translational Medicine, and Regulatory Science for the World's Rarest Conditions.","authors":"Cécile Ollivier, Solange Corriol-Rohou, Marta Del Álamo, Roseline Favresse, Johanna Kostenzer, Mathieu Boudes, Anton E Ussi, Klaus Viel, R Michael Linden, Magda Chlebus","doi":"10.1002/cpt.3428","DOIUrl":"https://doi.org/10.1002/cpt.3428","url":null,"abstract":"","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142034659","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Tyler Shugg, Emma M. Tillman, Amy M. Breman, Jennelle C. Hodge, Christine A. McDonald, Reynold C. Ly, Elizabeth J. Rowe, Wilberforce Osei, Tayler B. Smith, Peter H. Schwartz, John T. Callaghan, Victoria M. Pratt, Sheryl Lynch, Michael T. Eadon, Todd C. Skaar
In 2019, Indiana University launched the Precision Health Initiative to enhance the institutional adoption of precision medicine, including pharmacogenetics (PGx) implementation, at university-affiliated practice sites across Indiana. The overarching goal of this PGx implementation program was to facilitate the sustainable adoption of genotype-guided prescribing into routine clinical care. To accomplish this goal, we pursued the following specific objectives: (i) to integrate PGx testing into existing healthcare system processes; (ii) to implement drug–gene pairs with high-level evidence and educate providers and pharmacists on established clinical management recommendations; (iii) to engage key stakeholders, including patients to optimize the return of results for PGx testing; (iv) to reduce health disparities through the targeted inclusion of underrepresented populations; (v) and to track third-party reimbursement. This tutorial details our multifaceted PGx implementation program, including descriptions of our interventions, the critical challenges faced, and the major program successes. By describing our experience, we aim to assist other clinical teams in achieving sustainable PGx implementation in their health systems.
{"title":"Development of a Multifaceted Program for Pharmacogenetics Adoption at an Academic Medical Center: Practical Considerations and Lessons Learned","authors":"Tyler Shugg, Emma M. Tillman, Amy M. Breman, Jennelle C. Hodge, Christine A. McDonald, Reynold C. Ly, Elizabeth J. Rowe, Wilberforce Osei, Tayler B. Smith, Peter H. Schwartz, John T. Callaghan, Victoria M. Pratt, Sheryl Lynch, Michael T. Eadon, Todd C. Skaar","doi":"10.1002/cpt.3402","DOIUrl":"10.1002/cpt.3402","url":null,"abstract":"<p>In 2019, Indiana University launched the Precision Health Initiative to enhance the institutional adoption of precision medicine, including pharmacogenetics (PGx) implementation, at university-affiliated practice sites across Indiana. The overarching goal of this PGx implementation program was to facilitate the sustainable adoption of genotype-guided prescribing into routine clinical care. To accomplish this goal, we pursued the following specific objectives: (i) to integrate PGx testing into existing healthcare system processes; (ii) to implement drug–gene pairs with high-level evidence and educate providers and pharmacists on established clinical management recommendations; (iii) to engage key stakeholders, including patients to optimize the return of results for PGx testing; (iv) to reduce health disparities through the targeted inclusion of underrepresented populations; (v) and to track third-party reimbursement. This tutorial details our multifaceted PGx implementation program, including descriptions of our interventions, the critical challenges faced, and the major program successes. By describing our experience, we aim to assist other clinical teams in achieving sustainable PGx implementation in their health systems.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3402","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142015774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bonginkosi S'fiso Ndzamba, Samuel Egieyeh, Pius Fasinu
The availability of clinical trial data, advocacy, and increased funding has facilitated the implementation of pharmacometrics in Africa, resulting in the establishment of additional training programs for pharmacometricians. This study conducted a systematic review to evaluate the progress made from the implementation of pharmacometrics in clinical drug development and its adoption into drug approval by regulatory authorities in Africa. We performed a comprehensive literature search using major databases such as PubMed and Google Scholar. The study included articles published until 2024, with no lower cutoff. Articles were excluded if not addressing the research question or of pharmacometrics studies done outside Africa with no collaboration with African researchers (study setting). For the review, a total of 121 articles were included for analysis. Among the reported pharmacometrics approaches, Population pharmacokinetics modeling approaches are the most used (95 (78.5%)). South Africa and Uganda researchers have the most research output in pharmacometrics in Africa (82 (89.1%) and 7 (7.61%), respectively), with the University of Cape Town (South Africa) producing the highest (71 (78.8%)) of all article in Africa. The most studied conditions are TB (43 (35.5%)), HIV (33 (27.3%), TB and HIV (22 (18.2%)), and malaria (12 (9.92%). Pharmacometrics is gaining momentum in Africa, and the progress made since inception will significantly improve the safety and efficacy of therapeutic agents used to treat HIV, TB, and other emerging conditions.
{"title":"Progress in Pharmacometrics Implementation and Regulatory Integration in Africa: A Systematic Review.","authors":"Bonginkosi S'fiso Ndzamba, Samuel Egieyeh, Pius Fasinu","doi":"10.1002/cpt.3415","DOIUrl":"https://doi.org/10.1002/cpt.3415","url":null,"abstract":"<p><p>The availability of clinical trial data, advocacy, and increased funding has facilitated the implementation of pharmacometrics in Africa, resulting in the establishment of additional training programs for pharmacometricians. This study conducted a systematic review to evaluate the progress made from the implementation of pharmacometrics in clinical drug development and its adoption into drug approval by regulatory authorities in Africa. We performed a comprehensive literature search using major databases such as PubMed and Google Scholar. The study included articles published until 2024, with no lower cutoff. Articles were excluded if not addressing the research question or of pharmacometrics studies done outside Africa with no collaboration with African researchers (study setting). For the review, a total of 121 articles were included for analysis. Among the reported pharmacometrics approaches, Population pharmacokinetics modeling approaches are the most used (95 (78.5%)). South Africa and Uganda researchers have the most research output in pharmacometrics in Africa (82 (89.1%) and 7 (7.61%), respectively), with the University of Cape Town (South Africa) producing the highest (71 (78.8%)) of all article in Africa. The most studied conditions are TB (43 (35.5%)), HIV (33 (27.3%), TB and HIV (22 (18.2%)), and malaria (12 (9.92%). Pharmacometrics is gaining momentum in Africa, and the progress made since inception will significantly improve the safety and efficacy of therapeutic agents used to treat HIV, TB, and other emerging conditions.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142007916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Iris K. Minichmayr, Mohamad Shebley, Piet H. van der Graaf, Karthik Venkatakrishnan
Right dosage—administering the right dose at the right time in the optimal dosing interval using the appropriate application route and administration method—is central to the role of clinical pharmacology throughout the development and clinical use of therapeutics. It is critically important in all therapeutic areas to maximize patient benefit and minimize undesirable adverse effects. Getting the dosage right extends beyond population-level dosing to meet a certain efficacy or toxicity threshold in a defined patient group or subgroup of interest, such as patients with a specific indication or certain degree of organ impairment. As the “one-size-fits-all” concept often fails to optimize the benefit–risk ratio for all patients in clinical practice, various precision dosing strategies, acknowledging between-patient variability in exposure and/or response, have been evolving to tailor dosage regimens and increase the chances of treatment success for individual patients. Just as a sculptor meticulously chisels away at a block of marble to reveal a masterpiece, dose finding in drug development and precision dosing involves carefully tailoring dosage regimens to the disease of concern and the unique characteristics of patients (Figure 1). This special-themed issue of Clinical Pharmacology & Therapeutics illuminates various aspects, advancements, and future perspectives in getting the dosage right in drug development, regulatory approval, and clinical practice.
Dose selection and optimization have been particularly prominent topics in oncology in recent years, not least since the launch of Project Optimus by the US Food and Drug Administration.1 This initiative aims to reform dose selection to maximize not only the efficacy of a drug, but also its safety and tolerability. In this issue, two papers provide comprehensive viewpoints on the topic of oncology dosage optimization from the regulatory and pharmaceutical industry sectors, respectively. In their State-of-the-Art review, Rahman et al. offer a regulatory perspective highlighting the foundational importance of timely dosage optimization and the consequences of not doing so.2 The authors discuss the topic in the context of the realities of rapid development programs, rare and pediatric cancers, and combination development, outlining the value of tools in translational and precision medicine, and model-informed drug development for achieving the desired objectives. A White Paper by the Oncology Dose Optimization Working Group, commissioned by the International Consortium for Innovation and Quality in Pharmaceutical Development, highlights the impact of Project Optimus on oncology dose optimization, together with common issues and potential solutions, post-marketing requirements and commitments, as well as insights from a survey on current industry practices for oncology dose selection.3 The
{"title":"Getting the Dosage Right: A Vital Role for Clinical Pharmacology in the Era of Precision Medicine","authors":"Iris K. Minichmayr, Mohamad Shebley, Piet H. van der Graaf, Karthik Venkatakrishnan","doi":"10.1002/cpt.3375","DOIUrl":"10.1002/cpt.3375","url":null,"abstract":"<p>Right dosage—administering the right dose at the right time in the optimal dosing interval using the appropriate application route and administration method—is central to the role of clinical pharmacology throughout the development and clinical use of therapeutics. It is critically important in all therapeutic areas to maximize patient benefit and minimize undesirable adverse effects. Getting the dosage right extends beyond population-level dosing to meet a certain efficacy or toxicity threshold in a defined patient group or subgroup of interest, such as patients with a specific indication or certain degree of organ impairment. As the “one-size-fits-all” concept often fails to optimize the benefit–risk ratio for all patients in clinical practice, various precision dosing strategies, acknowledging between-patient variability in exposure and/or response, have been evolving to tailor dosage regimens and increase the chances of treatment success for <i>individual</i> patients. Just as a sculptor meticulously chisels away at a block of marble to reveal a masterpiece, dose finding in drug development and precision dosing involves carefully tailoring dosage regimens to the disease of concern and the unique characteristics of patients (<b>Figure</b> 1). This special-themed issue of <i>Clinical Pharmacology & Therapeutics</i> illuminates various aspects, advancements, and future perspectives in getting the dosage right in drug development, regulatory approval, and clinical practice.</p><p>Dose selection and optimization have been particularly prominent topics in oncology in recent years, not least since the launch of Project Optimus by the US Food and Drug Administration.<span><sup>1</sup></span> This initiative aims to reform dose selection to maximize not only the efficacy of a drug, but also its safety and tolerability. In this issue, two papers provide comprehensive viewpoints on the topic of oncology dosage optimization from the regulatory and pharmaceutical industry sectors, respectively. In their State-of-the-Art review, Rahman <i>et al</i>. offer a regulatory perspective highlighting the foundational importance of timely dosage optimization and the consequences of not doing so.<span><sup>2</sup></span> The authors discuss the topic in the context of the realities of rapid development programs, rare and pediatric cancers, and combination development, outlining the value of tools in translational and precision medicine, and model-informed drug development for achieving the desired objectives. A White Paper by the Oncology Dose Optimization Working Group, commissioned by the International Consortium for Innovation and Quality in Pharmaceutical Development, highlights the impact of Project Optimus on oncology dose optimization, together with common issues and potential solutions, post-marketing requirements and commitments, as well as insights from a survey on current industry practices for oncology dose selection.<span><sup>3</sup></span> The ","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3375","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142007918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Model-Informed Approaches to Optimizing Therapeutics in the African Patient Populations.","authors":"Mwila Mulubwa, Kelly Chibale","doi":"10.1002/cpt.3425","DOIUrl":"https://doi.org/10.1002/cpt.3425","url":null,"abstract":"","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142007914","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}