Thrombopoietin Human

Thrombopoietin Human Recombinant
Shipped with Ice Packs
In Stock

Description

TPO Human Recombinant produced in HEK293 cells is a single, glycosylated polypeptide chain containing 343 amino acids (22-353 a.a) and having a molecular mass of 36.8kDa.
TPO  is fused to a 6 amino acid His-tag at C-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Thrombopoietin, a glycoprotein hormone primarily produced by the liver and kidneys, plays a crucial role in regulating platelet production in the bone marrow. This hormone stimulates the production and differentiation of megakaryocytes, which are bone marrow cells that fragment into numerous platelets.
Description
Recombinant Human TPO, produced in HEK293 cells, is a single, glycosylated polypeptide chain consisting of 343 amino acids (22-353 a.a). It has a molecular mass of 36.8kDa and is fused to a 6 amino acid His-tag at the C-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless solution.
Formulation
The TPO protein solution has a concentration of 0.25mg/ml and contains 10% glycerol and Phosphate-Buffered Saline (pH 7.4).
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For longer periods, it is recommended to store the solution frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is greater than 90.0% as determined by SDS-PAGE analysis.
Biological Activity
The ED50 range, measured by a cell proliferation assay using MO7e human megakaryocytic leukemic cells, is approximately 10ng/ml.
Synonyms

Megakaryocyte colony-stimulating factor, Myeloproliferative leukemia virus oncogene ligand, C-mpl ligand, ML, Megakaryocyte growth and development factor, MGDF, TPO, MKCSF, MPLLG, MGC163194, THPO

Source

HEK293 cells.

Amino Acid Sequence

DGSHMSPAPP ACDLRVLSKL LRDSHVLHSR LSQCPEVHPL PTPVLLPAVD FSLGEWKTQM EETKAQDILG AVTLLLEGVM AARGQLGPTC LSSLLGQLSG QVRLLLGALQ SLLGTQLPPQ GRTTAHKDPN AIFLSFQHLL RGKVRFLMLV GGSTLCVRRA PPTTAVPSRT SLVLTLNELP NRTSGLLETN FTASARTTGS GLLKWQQGFR AKIPGLLNQT SRSLDQIPGY LNRIHELLNG TRGLFPGPSR RTLGAPDISS GTSDTGSLPP NLQPGYSPSP THPPTGQYTL FPLPPTLPTP VVQLHPLLPD PSAPTPTPTS PLLNTSYTHS QNLSQEGHHH HHH 

Q&A

What is recombinant human thrombopoietin (rhTPO) and how does it function?

Recombinant human thrombopoietin (rhTPO) is a full-length glycosylated cytokine produced by Chinese hamster ovary cells that mimics endogenous thrombopoietin, the primary physiological regulator of platelet production . It functions by binding to and activating the thrombopoietin receptor (MPL) on megakaryocytes and hematopoietic stem cells . This activation stimulates multiple downstream cellular pathways that promote the differentiation of bone marrow stem cells into megakaryocyte progenitor cells, induces the expression of megakaryocyte differentiation markers, and drives megakaryocyte proliferation and polyploidization, ultimately resulting in increased platelet formation in circulation .

The biological activities of rhTPO extend beyond platelet production, as it also plays potential roles in hematopoietic stem and progenitor cell regeneration, making it a candidate for investigation in various clinical contexts including relapsed immune thrombocytopenia, hematopoietic injury, and acute severe thrombocytopenia in sepsis .

How do researchers distinguish between rhTPO and other thrombopoietin receptor agonists?

Researchers distinguish between rhTPO and other thrombopoietin receptor agonists (TPO-RAs) based on several key characteristics:

  • Molecular structure: rhTPO is a full-length glycosylated protein highly similar to endogenous thrombopoietin, whereas other TPO-RAs like eltrombopag and romiplostim have different structures .

  • Mechanism of action: While all TPO-RAs activate the thrombopoietin receptor, they may do so through different binding sites or with different binding kinetics, leading to potential differences in downstream signaling .

  • Administration route: rhTPO is typically administered subcutaneously (e.g., 300U/kg daily), while eltrombopag is administered orally (starting at 25-50mg daily) .

  • Clinical response patterns: Studies directly comparing rhTPO with other agents, such as eltrombopag, evaluate differences in time to response, duration of response, and sustainability of platelet count elevation .

  • Safety profile: Different TPO-RAs may have distinct adverse effect profiles, including risks of thromboembolism, bone marrow fibrosis, or immunogenicity .

These distinctions are critically important for researchers designing clinical trials, as they inform patient selection, endpoint measurement, and safety monitoring protocols.

What methodologies are used to evaluate rhTPO efficacy in secondary failure of platelet recovery (SFPR)?

Researchers evaluating rhTPO efficacy in secondary failure of platelet recovery (SFPR) after allogeneic hematopoietic stem cell transplantation typically employ the following methodological approaches:

These methodologies allow researchers to comprehensively assess both the clinical efficacy and biological mechanisms of rhTPO in SFPR settings.

How are optimized dosing regimens for rhTPO determined in clinical research?

Determination of optimized rhTPO dosing regimens in clinical research follows a structured methodological approach:

  • Baseline stratification: Patients are stratified based on initial platelet counts (e.g., <20×10^9/L or ≥20-<30×10^9/L) to account for disease severity as a potential confounder in dose-response relationships .

  • Initial dose selection: Starting doses are typically determined from previous phase I studies, with common regimens including 300U/kg or 600U/kg subcutaneously once daily . This allows researchers to study different points on the dose-response curve.

  • Adaptive dosing protocol: Research protocols include pre-specified algorithms for dose adjustments based on individual platelet responses, with defined maximum doses (e.g., 600 U/kg/day for rhTPO) .

  • Pharmacokinetic/pharmacodynamic correlation: Serial measurements of plasma rhTPO levels are correlated with platelet response curves to establish optimal dosing intervals and maintenance regimens .

  • Endpoint measurement: Primary endpoints often include time to response (defined as time from treatment initiation to first achievement of platelet count ≥50×10^9/L), which directly assesses the speed of clinical efficacy at different doses .

  • Comparative design: Head-to-head comparisons with established therapies (such as eltrombopag) provide context for the clinical relevance of different dosing strategies .

  • Statistical modeling: Advanced statistical models are employed to identify dosing factors associated with optimal response while minimizing adverse effects, allowing for development of personalized dosing recommendations .

These systematic approaches ensure that dosing regimens balance maximal efficacy with patient safety and practical administration considerations.

What are the critical factors in designing clinical trials that evaluate rhTPO efficacy?

Designing robust clinical trials to evaluate rhTPO efficacy requires careful consideration of several critical factors:

  • Patient population selection:

    • Clear definition of the thrombocytopenic condition being studied (ITP, SFPR, etc.)

    • Appropriate inclusion/exclusion criteria to minimize confounding factors

    • Stratification based on relevant prognostic factors (e.g., baseline platelet count, disease duration)

  • Study design elements:

    • Control group selection (placebo or active comparator like eltrombopag)

    • Randomization method with appropriate allocation concealment

    • Blinding considerations (open-label vs. double-blind)

    • Treatment duration sufficient to capture both immediate and sustained responses

  • Endpoint selection and measurement:

    • Primary endpoints that reflect clinically meaningful outcomes (time to response, response rate)

    • Secondary endpoints capturing durability of response, bleeding events, quality of life

    • Standardized definitions of response (e.g., platelet count ≥50×10^9/L and at least 2-fold increase from baseline)

  • Safety monitoring protocols:

    • Systematic assessment of adverse events

    • Special attention to theoretical risks (thrombotic events, bone marrow fibrosis)

    • Immunogenicity monitoring for anti-TPO antibodies

  • Statistical considerations:

    • Appropriate sample size calculations based on expected effect sizes

    • Analysis plans that account for competing risks (e.g., death without reaching response)

    • Methods for handling missing data and early discontinuations

  • Subgroup analyses:

    • Pre-specified analyses of response in important patient subgroups

    • Biomarker studies to identify predictors of response or resistance

By addressing these factors systematically, researchers can generate high-quality evidence regarding rhTPO efficacy and safety across different clinical scenarios.

How can researchers effectively measure megakaryocyte response to rhTPO treatment?

Effective measurement of megakaryocyte response to rhTPO treatment requires a multifaceted approach combining morphological, functional, and molecular assessments:

  • Bone marrow morphological evaluation:

    • Quantification of total megakaryocyte (MK) numbers per high-power field

    • Classification of MKs into functional subtypes:

      • Thromocytogenic MKs (actively producing platelets)

      • Granular MKs (mature but not releasing platelets)

      • Immature MKs

    • Assessment of MK size, ploidy distribution, and morphological abnormalities

  • Statistical analysis of MK parameters:

    • Paired t-tests to compare pre- and post-treatment MK counts

    • Correlation analyses between MK metrics and peripheral platelet responses

    • Time-series analysis of MK recovery patterns

  • Flow cytometric analysis:

    • Measurement of megakaryocyte progenitor populations in bone marrow

    • Assessment of megakaryocyte maturation markers

    • Evaluation of platelet-producing capacity through proplatelet formation assays

  • Molecular profiling:

    • Gene expression analysis of megakaryocyte differentiation markers

    • Receptor expression studies to characterize TPO-R density and signaling

    • Transcriptomic profiling to identify mechanisms of response or resistance

One study documented significant increases in both total megakaryocytes (7.03 vs. 21.58, p=0.04) and thromocytogenic megakaryocytes (0.31 vs. 1.35, p=0.048) after rhTPO treatment, while granular megakaryocytes showed no significant increase (5.21 vs. 7.58, p=0.36) . These morphological changes correlated with progressive increases in peripheral platelet counts over time, providing mechanistic insights into the therapeutic effects of rhTPO.

This comprehensive approach allows researchers to dissect the cellular and molecular mechanisms by which rhTPO influences megakaryopoiesis and subsequent platelet production.

What factors predict clinical response to rhTPO therapy in different patient populations?

Predictive factors for clinical response to rhTPO therapy vary across patient populations and have been identified through multivariate analyses in several studies:

  • In secondary failure of platelet recovery (SFPR) after HSCT:

    • Higher CD34+ cell dose during transplantation is independently associated with better response to rhTPO (HR shown in multivariate analysis)

    • Particular underlying hematological malignancies may influence response patterns

    • Prior history of viral reactivation (CMV, EBV) affects response rates as time-dependent covariates

  • In immune thrombocytopenia (ITP):

    • Disease duration (chronic vs. newly diagnosed) impacts response rates

    • Prior treatment exposure patterns affect likelihood of response

    • Initial platelet count (<20×10^9/L vs. ≥20-<30×10^9/L) plays a role in stratification for response prediction

  • Across multiple indications:

    • Age appears to have variable impact on response, with some studies showing no significant effect of age ≥30 years vs. <30 years (HR 0.84, 95% CI 0.37-1.89, p=0.67)

    • HLA matching status in transplant settings does not significantly impact response (HR 0.97, 95% CI 0.44-2.18, p=0.95)

    • Use of anti-thymocyte globulin (ATG) during conditioning does not appear to significantly affect subsequent rhTPO response (HR 1.20, 95% CI 0.41-3.53, p=0.75)

  • Biological response predictors:

    • Baseline endogenous thrombopoietin levels (as in the case reports of pregnant ITP patients with levels of 231pg/mL and 107pg/mL before rhTPO therapy)

    • Bone marrow megakaryocyte patterns before treatment

    • Genetic polymorphisms in thrombopoietin receptor or signaling pathways

Understanding these predictive factors enables researchers to design more targeted studies and clinicians to select patients most likely to benefit from rhTPO therapy, improving research efficiency and clinical outcomes.

How do researchers analyze and interpret platelet response kinetics in rhTPO clinical trials?

Analysis and interpretation of platelet response kinetics in rhTPO clinical trials involves sophisticated methodological approaches:

These comprehensive analytical approaches allow researchers to characterize not only whether patients respond to rhTPO, but also the quality, timing, and durability of response—crucial factors for clinical decision-making and regulatory approval processes.

What are the methodological approaches to monitor and evaluate safety concerns with rhTPO therapy?

Monitoring and evaluating safety concerns with rhTPO therapy requires systematic methodological approaches:

  • Immunogenicity assessment:

    • Screening for neutralizing antibodies to rhTPO before and at regular intervals during treatment

    • In phase I studies, no neutralizing antibodies to rhTPO were observed, suggesting low immunogenicity risk

    • Correlation of antibody development with clinical response patterns

  • Bone marrow fibrosis monitoring:

    • Sequential bone marrow biopsies with standardized reticulin/collagen staining

    • Grading of fibrosis using validated scales

    • Studies of TPO receptor agonists indicate that while small numbers of patients develop moderate-severe reticulin fibrosis and/or collagen fibrosis, these changes are usually reversible upon discontinuation

  • Thrombotic risk evaluation:

    • Systematic monitoring for venous and arterial thromboembolism

    • Assessment of thrombotic risk factors at baseline and during treatment

    • Evidence indicates that TPO receptor agonists may increase venous thromboembolism risk

  • Concurrent medication effects:

    • Evaluation of interactions with other commonly used medications

    • Studies showing that rhTPO does not compromise neutrophil recovery when administered concurrently with G-CSF

  • Long-term safety surveillance:

    • Extended follow-up protocols beyond the primary treatment period

    • Monitoring for delayed adverse effects or secondary malignancies

    • Registry-based studies to capture rare adverse events in larger patient populations

  • Special population safety analyses:

    • Case reports and small series in pregnant women showing successful outcomes without apparent fetal complications

    • Dedicated pediatric safety studies

    • Safety profiles in patients with comorbidities

These methodological approaches ensure comprehensive safety evaluation of rhTPO across different clinical scenarios and patient populations, informing risk-benefit assessments for research and clinical applications.

What research methodologies are employed to study rhTPO in pregnant patients with thrombocytopenia?

Research on rhTPO in pregnant patients with thrombocytopenia employs specialized methodologies to address unique ethical and clinical considerations:

  • Case study design with detailed documentation:

    • Individual case reports with comprehensive documentation of maternal and fetal parameters

    • Standardized reporting of gestational age, disease characteristics, prior treatments, and rhTPO administration details

    • Example: Two pregnant ITP patients (a 28-year-old G2P1 woman and a 23-year-old primigravid woman) who failed to respond to prednisone and IVIG were treated with rhTPO 300U/kg daily until platelet counts exceeded 50×10^9/L, then switched to maintenance therapy (rhTPO 300U/kg every other day)

  • Baseline assessment protocol:

    • Documentation of pre-treatment platelet count (e.g., 10×10^9/L with gingival bleeding in one patient; 19×10^9/L with gross hematuria at 22-week gestation in another)

    • Measurement of endogenous thrombopoietin levels before treatment (231pg/mL and 107pg/mL in the reported cases)

    • Careful assessment of bleeding manifestations and transfusion requirements

  • Response monitoring framework:

    • Serial platelet count measurements at standardized intervals

    • One patient's count rose to 165×10^9/L after 10 days of rhTPO therapy

    • Assessment of bleeding cessation and prevention of recurrence

  • Maternal-fetal outcome evaluation:

    • Documentation of delivery method, gestational age at delivery, and neonatal outcomes

    • Assessment of neonatal platelet counts to evaluate for potential transplacental effects

    • One patient vaginally delivered a female infant with normal platelet count at 39 weeks

  • Ethical and regulatory framework:

    • Approval by Medical Ethical Committees (e.g., Qilu Hospital, Shandong University)

    • Informed consent with specific documentation of risk-benefit considerations in pregnancy

    • Transparent reporting of decision-making process for using experimental therapy in this vulnerable population

These specialized methodological approaches provide valuable preliminary data on rhTPO use in pregnancy while addressing the ethical complexities of research in this population, for whom few established treatment options exist when conventional therapies fail.

What methodological approaches are used to compare rhTPO with other thrombopoietin receptor agonists?

Comparative research between rhTPO and other thrombopoietin receptor agonists (TPO-RAs) employs several sophisticated methodological approaches:

  • Randomized controlled trial design:

    • Multicenter, open-label randomized controlled trials with specified allocation ratios (e.g., 2:1 for rhTPO vs. eltrombopag)

    • Stratification by key prognostic factors such as baseline platelet count (<20×10^9/L or ≥20-<30×10^9/L)

    • Intention-to-treat analysis for primary efficacy endpoints

  • Standardized intervention protocols:

    • Clear definition of initial doses (e.g., rhTPO at 300U/kg or 600U/kg subcutaneously once daily vs. eltrombopag at 25mg or 50mg orally once daily)

    • Predefined dose adjustment algorithms based on platelet response

    • Specified maximum doses (e.g., 600 U/kg/day for rhTPO and 75 mg/day for eltrombopag)

  • Endpoint harmonization:

    • Primary endpoints focused on time-based metrics (e.g., time to response defined as time from treatment initiation to first achievement of platelet count ≥50×10^9/L)

    • Secondary endpoints including response rates (platelet count ≥30×10^9/L and at least 2-fold increase from baseline without bleeding)

    • Durability measures such as duration of platelet count maintenance ≥50×10^9/L and time to treatment failure

  • Comprehensive safety assessment:

    • Standardized adverse event reporting

    • Systematic monitoring for class-specific concerns (thrombosis, bone marrow fibrosis)

    • Immunogenicity testing for anti-TPO antibodies

  • Cost-effectiveness analysis:

    • Calculation of cost per responder

    • Healthcare resource utilization metrics (hospitalization, bleeding-related interventions)

    • Quality-adjusted life year (QALY) assessments

The first comparative study between optimized rhTPO regimens and standard eltrombopag provides valuable methodological frameworks for future comparative effectiveness research in this area , establishing standards for balanced assessment of these therapeutic options across multiple dimensions of efficacy, safety, and resource utilization.

How do researchers account for pharmacological differences when comparing various thrombopoietin receptor agonists?

Researchers employ specialized methodological approaches to account for pharmacological differences when comparing thrombopoietin receptor agonists:

  • Pharmacokinetic/pharmacodynamic (PK/PD) modeling:

    • Characterization of absorption profiles (rhTPO is administered subcutaneously while eltrombopag is given orally)

    • Determination of elimination half-lives and volume of distribution for each agent

    • Development of mathematical models correlating plasma drug levels with platelet response

  • Dose normalization techniques:

    • Establishment of biologically equivalent doses based on receptor occupancy

    • Utilization of maximum effective doses for each agent (600 U/kg/day for rhTPO, 75 mg/day for eltrombopag)

    • Allowance for individualized dose titration within predefined algorithms to account for variable dose-response relationships

  • Receptor binding and signaling studies:

    • Comparative analysis of binding sites on the thrombopoietin receptor

    • Assessment of differences in downstream signaling pathway activation

    • Evaluation of receptor internalization and recycling dynamics

  • Time-matched response assessments:

    • Standardized assessment timepoints regardless of pharmacokinetic differences

    • Time-to-response analyses that account for expected pharmacological onset differences

    • Area-under-the-curve analyses for platelet count over time

  • Mechanistic biomarker evaluation:

    • Measurement of megakaryocyte numbers and morphology changes with different agents

    • Assessment of platelet function in addition to platelet count

    • Evaluation of effects on other hematopoietic lineages

  • Route of administration considerations:

    • Accounting for compliance differences between oral (eltrombopag) and injectable (rhTPO) formulations

    • Evaluation of convenience factors and their impact on long-term adherence

    • Analysis of administration-related adverse events

These methodological approaches ensure that comparative studies account for fundamental pharmacological differences between agents, allowing for fair and clinically relevant comparisons that inform research directions and clinical decision-making.

What novel applications of rhTPO are being investigated beyond current indications?

Researchers are exploring several novel applications for rhTPO beyond its established indications:

  • Hematopoietic stem and progenitor cell regeneration:

    • Investigation of rhTPO's potential to stimulate broader hematopoietic recovery

    • Exploration of mechanisms beyond direct thrombopoiesis

    • Applications in radiation injury, chemotherapy-induced bone marrow suppression, and other forms of marrow failure

  • Acute severe thrombocytopenia in sepsis:

    • Research into the pathophysiology of sepsis-associated thrombocytopenia

    • Evaluation of rhTPO's ability to counteract sepsis-induced thrombopoietic suppression

    • Assessment of impact on bleeding complications and mortality in septic patients

  • Combination therapies for refractory conditions:

    • Studies of rhTPO in combination with other thrombopoietic agents or immunomodulators

    • Evaluation of synergistic effects with complementary mechanisms of action

    • Development of sequential therapy protocols to maximize response rates

  • Surgical and procedural thrombocytopenia management:

    • Preoperative platelet augmentation to reduce transfusion requirements

    • Protocol development for patients requiring invasive procedures who cannot receive platelet transfusions

    • Risk-benefit assessments in settings where rapid but temporary platelet increase is needed

  • Ex vivo platelet generation:

    • Utilization of rhTPO in bioreactor systems for platelet production from donor stem cells

    • Development of protocols for generating functional platelets for transfusion

    • Research into extension of platelet shelf-life and functionality

  • Hepatitis-associated thrombocytopenia:

    • Building on eltrombopag's existing indication in hepatitis C

    • Investigation of rhTPO's specific advantages in liver disease settings

    • Management of thrombocytopenia to enable antiviral therapy completion

These emerging research directions reflect a growing understanding of thrombopoietin biology and the potential therapeutic applications of rhTPO beyond its initial development focus, representing promising areas for future clinical investigation.

What methodological challenges exist in advancing rhTPO research, and how might they be addressed?

Advancing rhTPO research faces several methodological challenges that require innovative approaches:

  • Long-term safety assessment:

    • Challenge: Limited data on extended rhTPO administration beyond clinical trials

    • Solution: Establishment of patient registries with standardized follow-up protocols for 5+ years

    • Approach: Implementation of systematic bone marrow monitoring and thrombotic event surveillance in routine clinical practice

  • Biomarker development for response prediction:

    • Challenge: Difficulty identifying reliable predictors of rhTPO response

    • Solution: Integration of -omics approaches (genomics, proteomics, metabolomics) with clinical data

    • Approach: Prospective biobanking of samples from all clinical trials with standardized processing and analysis protocols

  • Optimal combination therapy protocols:

    • Challenge: Numerous potential combinations with limited resources for testing

    • Solution: Adaptive trial designs that allow rapid testing of multiple combinations

    • Approach: Bayesian statistical methods to identify promising combinations early in the developmental pipeline

  • Resistance mechanisms identification:

    • Challenge: Understanding why some patients fail to respond or lose response

    • Solution: Sequential sampling of non-responders for biological characterization

    • Approach: Single-cell RNA sequencing of megakaryocyte populations before and during treatment

  • Special population dosing optimization:

    • Challenge: Limited data in pregnant patients, pediatric populations, and those with organ dysfunction

    • Solution: Physiologically-based pharmacokinetic modeling to predict appropriate dosing

    • Approach: Careful case series with intensive pharmacokinetic sampling to validate models

  • Comparative effectiveness research standardization:

    • Challenge: Heterogeneity in endpoints across studies of different TPO-RAs

    • Solution: Development of core outcome sets through international consensus

    • Approach: Implementation of patient-relevant outcomes alongside traditional hematologic parameters

  • Cost-effectiveness assessment:

    • Challenge: Limited economic data comparing rhTPO with alternatives

    • Solution: Integration of economic endpoints in clinical trials

    • Approach: Development of disease-specific quality of life instruments sensitive to thrombocytopenia-related morbidity

By addressing these methodological challenges through innovative research approaches, investigators can advance the field of rhTPO research and optimize its application across various clinical scenarios, ultimately improving outcomes for patients with thrombocytopenic disorders.

What are the key takeaways from current rhTPO research for clinical investigators?

The current body of research on recombinant human thrombopoietin provides several important takeaways for clinical investigators:

These evidence-based takeaways provide a foundation for clinical investigators designing future studies and for clinicians incorporating rhTPO into treatment algorithms for patients with thrombocytopenic disorders.

How should researchers interpret the cumulative evidence on rhTPO in planning future studies?

Researchers planning future studies on rhTPO should interpret the cumulative evidence through several critical lenses:

  • Gaps in current knowledge:

    • Limited data on long-term safety beyond initial clinical trials

    • Insufficient understanding of resistance mechanisms and predictors of response

    • Need for optimization of combination strategies with other agents

    • Limited exploration in certain patient subgroups (pediatric, elderly, organ dysfunction)

  • Methodological strengths to build upon:

    • Well-established response criteria facilitating cross-study comparisons

    • Validated safety monitoring approaches for bone marrow fibrosis and thrombotic events

    • Evolving understanding of optimal dosing strategies and pharmacokinetics

  • Design considerations for future trials:

    • Inclusion of biomarker studies to identify response predictors

    • Longer follow-up periods to assess durability of response and late effects

    • Incorporation of quality-of-life and functional measures beyond platelet counts

    • Consideration of pragmatic trial designs to assess real-world effectiveness

  • Comparative effectiveness priorities:

    • Head-to-head comparisons with other TPO-RAs using standardized endpoints

    • Cost-effectiveness analyses incorporating both direct and indirect costs

    • Patient preference studies to understand treatment burden and adherence factors

  • Translational research opportunities:

    • Investigation of molecular mechanisms underlying variable response

    • Exploration of effects on hematopoietic stem cell compartment beyond megakaryopoiesis

    • Development of ex vivo systems to predict in vivo response

  • Regulatory and policy implications:

    • Consideration of endpoints most relevant to regulatory approval

    • Development of risk minimization strategies based on cumulative safety data

    • Attention to healthcare system implementation factors

Product Science Overview

Introduction

Thrombopoietin (TPO) is a crucial glycoprotein hormone primarily responsible for the regulation of platelet production in the body. It plays a significant role in hematopoiesis, particularly in the development of megakaryocytes, the bone marrow cells that produce platelets. Recombinant human thrombopoietin (rhTPO) is a synthetic form of this hormone, engineered to mimic the natural TPO and used in various clinical applications.

Discovery and Purification

The journey of TPO began with its purification in 1994. This milestone led to the development of two recombinant forms of the c-Mpl ligand: recombinant human thrombopoietin (rhTPO) and pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) . These recombinant forms have undergone extensive clinical investigations to evaluate their efficacy and safety.

Biological Function

TPO binds to and activates the TPO receptor (c-Mpl) on megakaryocytes and their progenitors, promoting their growth and differentiation into platelets . This process is vital for maintaining adequate platelet levels in the blood, which are essential for normal blood clotting and wound healing.

Clinical Applications

Recombinant human thrombopoietin has shown promise in several clinical settings:

  1. Thrombocytopenia Management: Thrombocytopenia, a condition characterized by low platelet counts, is a common issue in patients undergoing chemotherapy, stem cell transplantation, and those with chronic liver disease or immune thrombocytopenia (ITP). RhTPO has been effective in reducing thrombocytopenia in these patients .

  2. Pregnancy: RhTPO has emerged as a novel therapeutic option for managing immune thrombocytopenia in pregnant women. Clinical studies have reported successful outcomes in increasing platelet counts and reducing bleeding risks in such patients .

  3. Other Potential Uses: Animal studies suggest that TPO may be beneficial in reducing surgical thrombocytopenia and bleeding, expanding pluripotent stem cells ex vivo, and serving as a radioprotectant .

Safety and Efficacy

While rhTPO has demonstrated significant potential, its use is not without challenges. Clinical trials have shown that rhTPO is a potent stimulator of megakaryocyte growth and platelet production. However, its benefits in stem cell transplantation and leukemia chemotherapy remain inconclusive . Ongoing and future studies are essential to fully understand the clinical role of rhTPO and its long-term safety.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.