search
Back to results

Intra-patient Dose Escalation Study to Investigate Safety and Feasibility of Vactosertib in Treating Anemic MPN Patients

Primary Purpose

Myeloproliferative Neoplasm

Status
Recruiting
Phase
Phase 2
Locations
United States
Study Type
Interventional
Intervention
Vactosertib
Sponsored by
Weill Medical College of Cornell University
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Myeloproliferative Neoplasm focused on measuring Anemia

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

Patients who meet the WHO 2016 criteria for a Ph-neg MPN (including PV, ET, MF, MDS/MPN, MPN-U).

  • Patients with MF must have DIPSS+ Intermediate or High-risk MF (primary of post-PV/ET).
  • Anemia as defined by HGB < 10 g/dL, or transfusion of ≥ 2 packed red blood cell (PRBC) unit within the past 4 weeks with HGB ≤8.5g/dL.
  • Ineligible, unsuitable or refractoriness to ESA therapy defined as any of the following:

    • Serum erythropoietin (EPO) >125 U/L.
    • Proven ESA unsuitability is defined by history of any of the following:
    • Loss of erythroid hematologic improvement while receiving stable or increased ESA dose; or
    • ESA-attributed toxicity that, in the treating physician's opinion, makes ESA therapy unsuitable for subject.
    • ESA refractoriness defined by lack of erythroid hematologic improvement to ESA:27
    • Less than 1.5 g/dL increase in hemoglobin after at least 6 weeks of ESA therapy; or
    • Ongoing transfusion dependence that has not been reduced by > 4U over an 8-week period compared to ESA pre-treatment 8 weeks.
  • Acceptable Cardiovascular status

Exclusion Criteria:

  • Any other serious medical condition which in the Investigator's opinion would preclude safe participation in the study.
  • Patients with history of TIA or stroke within the past 12 months are excluded.
  • Female subjects who are breastfeeding, or intend to breastfeed, during the study or in the 30 days following the last dose of study drug are excluded.

Sites / Locations

  • Weill Medical College of Cornell UniversityRecruiting

Arms of the Study

Arm 1

Arm Type

Experimental

Arm Label

Treatment arm

Arm Description

Vactosertib intra-patient dose finding cohort.

Outcomes

Primary Outcome Measures

Number of adverse events
To establish a safe and tolerable dose and schedule of vactosertib in Philadelphia chromosome negative Myeloproliferative Neoplasms by recording all adverse events in patients receiving any dose of the drug. All adverse events will be documented accurately regardless of relationship to the study drug. The investigators will assess each adverse event and determine relatedness to study drug.
Change in symptoms of the disease while taking vactosertib
To assess the efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring symptomatic response. Symptomatic responses will be recorded as the change in scores on the Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score (MPN-SAF-TSS) from baseline through to the end of study. The MPN-SAF-TSS is a questionnaire with questions related to different symptoms that are graded from 0 (not present) through 10 (worst imaginable). The scores are calculated by adding the score for each question at each time the questionnaire is completed. All scores will be compared to the score that was calculated at baseline to determine if there was a symptom response. A symptom response is defined as a change in score by more than 50% decrease from baseline.
Change in spleen size while taking vactosertib
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring splenic response. Splenic response will be measured by the change from baseline in spleen volume on a sonogram of the upper left quadrant (at the end of Tier treatment) and in the change from baseline of the spleen length measured by palpation (at each visit until the end of study).
Change in transfusion dependency while taking vactosertib
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring erythropoietic response. Erythropoietic response will be measured by the change from baseline of transfusion dependency.This will be measured by comparing the number of transfusions a subject required in the 8 weeks prior to beginning therapy to the number of transfusions required while on study.
Change in hemoglobin values while taking vactosertib
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring erythropoietic response. Erythropoietic response will be measured by the change in hemoglobin values from baseline. This will be measured by obtaining Complete Blood Counts (CBC) at every study visit.
Change in EPO levels while taking vactosertib
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring erythropoietic response. Erythropoietic response will be measured by the change in EPO levels from baseline. Serum EPO levels will be measured on day 1 of each cycle.

Secondary Outcome Measures

Change in MPN driver mutation ratios in patients taking vactosertib
To evaluate the on target molecular activity of vactosertib in anemic patients with Philadelphia negative Myeloproloferative Neoplasms. This will be determined by measuring the change in blood (and/or marrow) allelic ratio of MPN driver mutations (JAK2, CALR or MPL).
Histologic change in the bone marrow
To evaluate the on target molecular activity of vactosertib in anemic patients with Philadelphia negative Myeloproloferative Neoplasms. This will be measured by the histologic response patients have to vactosertib. Histologic response will be measured by change in bone marrow biopsy cellularity and fibrosis grade.
Change in Molecular activity of vactosertib
To evaluate the on target molecular activity of vactosertib in anemic patients with Philadelphia negative Myeloproloferative Neoplasms. This will be measured by looking at the SMAD2/3 phosphorylation measured by flow cytometry of peripheral blood and/or bone marrow hematopoietic cells or by immunohistochemical staining of bone marrow biopsy sections.

Full Information

First Posted
September 18, 2019
Last Updated
December 19, 2022
Sponsor
Weill Medical College of Cornell University
search

1. Study Identification

Unique Protocol Identification Number
NCT04103645
Brief Title
Intra-patient Dose Escalation Study to Investigate Safety and Feasibility of Vactosertib in Treating Anemic MPN Patients
Official Title
A 2-tiered, Phase 2, Rule-based, Intra-patient Dose Escalation Study to Investigate Safety and Feasibility of Vactosertib (TEW-7197) in the Treatment of Anemic Patients With Philadelphia Chromosome-negative MPNs (Ph-neg MPNs)
Study Type
Interventional

2. Study Status

Record Verification Date
December 2022
Overall Recruitment Status
Recruiting
Study Start Date
November 22, 2019 (Actual)
Primary Completion Date
September 1, 2023 (Anticipated)
Study Completion Date
September 1, 2024 (Anticipated)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Weill Medical College of Cornell University

4. Oversight

Studies a U.S. FDA-regulated Drug Product
Yes
Studies a U.S. FDA-regulated Device Product
No
Data Monitoring Committee
Yes

5. Study Description

Brief Summary
This study assesses the potential of using a TGFβ receptor inhibitor for the treatment of anemic patients with myeloproliferetive neoplasms. TGFβ signaling is known to be abnormally high in patients with myeloproliferative neoplasms and it is thought that abnormal TGFβ signals cause many of the problems with blood cell formation in these diseases. The study design allows all patients to receive the study drug, vactosertib. The dose of vactosertib is individualized within a pre-set range based upon its effectiveness and tolerability. A total of up to 37 patients will be treated.
Detailed Description
This is a two-tiered single arm Phase 2 trial of vactosertib (TEW-7197) for the treatment of anemia in Ph-neg MPNs. Both tiers use a rule-based, accelerated dose escalation scheme to efficiently assess the potential of vactosertib to safely and effectively treat anemic patients with Ph-neg MPNs. The first tier of this trial (Tier 1) is an intra-patient dose finding study in 12 patients that uses a low starting dose of vactosertib for all patients. For each patent, the treatment dose is escalated according to prospectively-defined rules, and a toxicity and treatment effect algorithm during the period of 16 weeks (4 treatment cycles). If pre-established efficacy and safety endpoints are met (section 5.4, section 9.1, section 11.1), then Tier 1 of the study will be followed by a Tier 2 expansion study with an additional 25 patients for a period of 24 weeks (6 treatment cycles). Vactosertib will be administered as monotherapy and therefore patients must be off cytoreductive therapies such as interferon, ruxolitinib, hydroxyurea, DNA hypomethylating agents or other cytotoxic chemotherapy prior to enrollment for a period of at least 14 days or 5 half-lives, whichever is longer. Supportive care measures including packed red blood cell (PRBC) transfusions for HGB <7g/dL, or symptomatic anemia, will be permitted. Administration of erythropoiesis stimulating agents (ESAs), however, will not be permitted on the trial (patients recruited would have serum EPO >125 U/L above which the benefit of ESAs is not supported).

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Myeloproliferative Neoplasm
Keywords
Anemia

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 2
Interventional Study Model
Single Group Assignment
Model Description
This is an intra-patient dose finding study which starts with low dose of vactosertib.
Masking
None (Open Label)
Allocation
N/A
Enrollment
37 (Anticipated)

8. Arms, Groups, and Interventions

Arm Title
Treatment arm
Arm Type
Experimental
Arm Description
Vactosertib intra-patient dose finding cohort.
Intervention Type
Drug
Intervention Name(s)
Vactosertib
Other Intervention Name(s)
TEW-7197
Intervention Description
This drug is a TGF-Beta receptor type 1 inhibitor, by inhibiting phosphorylation of the ALK5 substrates SMAD2 and SMAD3. This inhibition could promote regeneration of normal human stem cells and proliferation of erythroid progenitors to treat the underlying hypoproliferative anemia in advanced MPNs.
Primary Outcome Measure Information:
Title
Number of adverse events
Description
To establish a safe and tolerable dose and schedule of vactosertib in Philadelphia chromosome negative Myeloproliferative Neoplasms by recording all adverse events in patients receiving any dose of the drug. All adverse events will be documented accurately regardless of relationship to the study drug. The investigators will assess each adverse event and determine relatedness to study drug.
Time Frame
16 weeks
Title
Change in symptoms of the disease while taking vactosertib
Description
To assess the efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring symptomatic response. Symptomatic responses will be recorded as the change in scores on the Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score (MPN-SAF-TSS) from baseline through to the end of study. The MPN-SAF-TSS is a questionnaire with questions related to different symptoms that are graded from 0 (not present) through 10 (worst imaginable). The scores are calculated by adding the score for each question at each time the questionnaire is completed. All scores will be compared to the score that was calculated at baseline to determine if there was a symptom response. A symptom response is defined as a change in score by more than 50% decrease from baseline.
Time Frame
From baseline through 40 weeks
Title
Change in spleen size while taking vactosertib
Description
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring splenic response. Splenic response will be measured by the change from baseline in spleen volume on a sonogram of the upper left quadrant (at the end of Tier treatment) and in the change from baseline of the spleen length measured by palpation (at each visit until the end of study).
Time Frame
From baseline through 40 weeks
Title
Change in transfusion dependency while taking vactosertib
Description
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring erythropoietic response. Erythropoietic response will be measured by the change from baseline of transfusion dependency.This will be measured by comparing the number of transfusions a subject required in the 8 weeks prior to beginning therapy to the number of transfusions required while on study.
Time Frame
From baseline through 40 weeks
Title
Change in hemoglobin values while taking vactosertib
Description
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring erythropoietic response. Erythropoietic response will be measured by the change in hemoglobin values from baseline. This will be measured by obtaining Complete Blood Counts (CBC) at every study visit.
Time Frame
From baseline through 40 weeks
Title
Change in EPO levels while taking vactosertib
Description
To assess efficacy of vactosertib in treating anemic patients with Philadelphia chromosome negative Myeloproliferative Neoplasms by measuring erythropoietic response. Erythropoietic response will be measured by the change in EPO levels from baseline. Serum EPO levels will be measured on day 1 of each cycle.
Time Frame
From baseline through 40 weeks
Secondary Outcome Measure Information:
Title
Change in MPN driver mutation ratios in patients taking vactosertib
Description
To evaluate the on target molecular activity of vactosertib in anemic patients with Philadelphia negative Myeloproloferative Neoplasms. This will be determined by measuring the change in blood (and/or marrow) allelic ratio of MPN driver mutations (JAK2, CALR or MPL).
Time Frame
From baseline through 40 weeks
Title
Histologic change in the bone marrow
Description
To evaluate the on target molecular activity of vactosertib in anemic patients with Philadelphia negative Myeloproloferative Neoplasms. This will be measured by the histologic response patients have to vactosertib. Histologic response will be measured by change in bone marrow biopsy cellularity and fibrosis grade.
Time Frame
From baseline through 40 weeks
Title
Change in Molecular activity of vactosertib
Description
To evaluate the on target molecular activity of vactosertib in anemic patients with Philadelphia negative Myeloproloferative Neoplasms. This will be measured by looking at the SMAD2/3 phosphorylation measured by flow cytometry of peripheral blood and/or bone marrow hematopoietic cells or by immunohistochemical staining of bone marrow biopsy sections.
Time Frame
From baseline through 40 weeks

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: Patients who meet the WHO 2016 criteria for a Ph-neg MPN (including PV, ET, MF, MDS/MPN, MPN-U). Patients with MF must have DIPSS+ Intermediate or High-risk MF (primary of post-PV/ET). Anemia as defined by HGB < 10 g/dL, or transfusion of ≥ 2 packed red blood cell (PRBC) unit within the past 4 weeks with HGB ≤8.5g/dL. Ineligible, unsuitable or refractoriness to ESA therapy defined as any of the following: Serum erythropoietin (EPO) >125 U/L. Proven ESA unsuitability is defined by history of any of the following: Loss of erythroid hematologic improvement while receiving stable or increased ESA dose; or ESA-attributed toxicity that, in the treating physician's opinion, makes ESA therapy unsuitable for subject. ESA refractoriness defined by lack of erythroid hematologic improvement to ESA:27 Less than 1.5 g/dL increase in hemoglobin after at least 6 weeks of ESA therapy; or Ongoing transfusion dependence that has not been reduced by > 4U over an 8-week period compared to ESA pre-treatment 8 weeks. Acceptable Cardiovascular status Exclusion Criteria: Any other serious medical condition which in the Investigator's opinion would preclude safe participation in the study. Patients with history of TIA or stroke within the past 12 months are excluded. Female subjects who are breastfeeding, or intend to breastfeed, during the study or in the 30 days following the last dose of study drug are excluded.
Central Contact Person:
First Name & Middle Initial & Last Name or Official Title & Degree
Joseph M Scandura, MD, PhD
Phone
646-962-2700
Email
jms2003@med.cornell.edu
First Name & Middle Initial & Last Name or Official Title & Degree
Niamh Savage
Phone
212-746-1493
Email
nis2049@med.cornell.edu
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Joseph M Scandura, MD, PhD
Organizational Affiliation
Weill Medical College of Cornell University
Official's Role
Principal Investigator
Facility Information:
Facility Name
Weill Medical College of Cornell University
City
New York
State/Province
New York
ZIP/Postal Code
10021
Country
United States
Individual Site Status
Recruiting
Facility Contact:
First Name & Middle Initial & Last Name & Degree
Joseph Scandura, MD, PhD
Phone
212-746-6736
Email
jms2003@med.cornell.edu
First Name & Middle Initial & Last Name & Degree
Niamh Savage
Phone
212-746-1493

12. IPD Sharing Statement

Plan to Share IPD
No
Citations:
PubMed Identifier
28009442
Citation
Hernandez-Boluda JC, Correa JG, Garcia-Delgado R, Martinez-Lopez J, Alvarez-Larran A, Fox ML, Garcia-Gutierrez V, Perez-Encinas M, Ferrer-Marin F, Mata-Vazquez MI, Raya JM, Estrada N, Garcia S, Kerguelen A, Duran MA, Albors M, Cervantes F. Predictive factors for anemia response to erythropoiesis-stimulating agents in myelofibrosis. Eur J Haematol. 2017 Apr;98(4):407-414. doi: 10.1111/ejh.12846. Epub 2017 Jan 19.
Results Reference
background
PubMed Identifier
23838352
Citation
Tefferi A, Cervantes F, Mesa R, Passamonti F, Verstovsek S, Vannucchi AM, Gotlib J, Dupriez B, Pardanani A, Harrison C, Hoffman R, Gisslinger H, Kroger N, Thiele J, Barbui T, Barosi G. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. Blood. 2013 Aug 22;122(8):1395-8. doi: 10.1182/blood-2013-03-488098. Epub 2013 Jul 9.
Results Reference
background
PubMed Identifier
23591792
Citation
Barosi G, Mesa R, Finazzi G, Harrison C, Kiladjian JJ, Lengfelder E, McMullin MF, Passamonti F, Vannucchi AM, Besses C, Gisslinger H, Samuelsson J, Verstovsek S, Hoffman R, Pardanani A, Cervantes F, Tefferi A, Barbui T. Revised response criteria for polycythemia vera and essential thrombocythemia: an ELN and IWG-MRT consensus project. Blood. 2013 Jun 6;121(23):4778-81. doi: 10.1182/blood-2013-01-478891. Epub 2013 Apr 16.
Results Reference
background
PubMed Identifier
25037629
Citation
Tefferi A, Guglielmelli P, Larson DR, Finke C, Wassie EA, Pieri L, Gangat N, Fjerza R, Belachew AA, Lasho TL, Ketterling RP, Hanson CA, Rambaldi A, Finazzi G, Thiele J, Barbui T, Pardanani A, Vannucchi AM. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood. 2014 Oct 16;124(16):2507-13; quiz 2615. doi: 10.1182/blood-2014-05-579136. Epub 2014 Jul 18.
Results Reference
background
PubMed Identifier
25616577
Citation
Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, Winton EF, Harvey JH Jr, Arcasoy MO, Hexner EO, Lyons RM, Raza A, Vaddi K, Sun W, Peng W, Sandor V, Kantarjian H; COMFORT-I investigators. Efficacy, safety, and survival with ruxolitinib in patients with myelofibrosis: results of a median 3-year follow-up of COMFORT-I. Haematologica. 2015 Apr;100(4):479-88. doi: 10.3324/haematol.2014.115840. Epub 2015 Jan 23.
Results Reference
background
PubMed Identifier
28674026
Citation
Newberry KJ, Patel K, Masarova L, Luthra R, Manshouri T, Jabbour E, Bose P, Daver N, Cortes J, Kantarjian H, Verstovsek S. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood. 2017 Aug 31;130(9):1125-1131. doi: 10.1182/blood-2017-05-783225. Epub 2017 Jul 3.
Results Reference
background
PubMed Identifier
28860323
Citation
Vannucchi AM, Guglielmelli P. Traffic lights for ruxolitinib. Blood. 2017 Aug 31;130(9):1075-1077. doi: 10.1182/blood-2017-07-795880. No abstract available.
Results Reference
background
PubMed Identifier
19366369
Citation
Huang J, Tefferi A. Erythropoiesis stimulating agents have limited therapeutic activity in transfusion-dependent patients with primary myelofibrosis regardless of serum erythropoietin level. Eur J Haematol. 2009 Aug;83(2):154-5. doi: 10.1111/j.1600-0609.2009.01266.x. Epub 2009 Apr 10. No abstract available.
Results Reference
background
PubMed Identifier
12968110
Citation
Akel S, Petrow-Sadowski C, Laughlin MJ, Ruscetti FW. Neutralization of autocrine transforming growth factor-beta in human cord blood CD34(+)CD38(-)Lin(-) cells promotes stem-cell-factor-mediated erythropoietin-independent early erythroid progenitor development and reduces terminal differentiation. Stem Cells. 2003;21(5):557-67. doi: 10.1634/stemcells.21-5-557.
Results Reference
background
PubMed Identifier
19796233
Citation
Soderberg SS, Karlsson G, Karlsson S. Complex and context dependent regulation of hematopoiesis by TGF-beta superfamily signaling. Ann N Y Acad Sci. 2009 Sep;1176:55-69. doi: 10.1111/j.1749-6632.2009.04569.x.
Results Reference
background
PubMed Identifier
15477587
Citation
Scandura JM, Boccuni P, Massague J, Nimer SD. Transforming growth factor beta-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation. Proc Natl Acad Sci U S A. 2004 Oct 19;101(42):15231-6. doi: 10.1073/pnas.0406771101. Epub 2004 Oct 11. Erratum In: Proc Natl Acad Sci U S A. 2004 Nov 23;101(47):16707.
Results Reference
background
PubMed Identifier
23440043
Citation
Brenet F, Kermani P, Spektor R, Rafii S, Scandura JM. TGFbeta restores hematopoietic homeostasis after myelosuppressive chemotherapy. J Exp Med. 2013 Mar 11;210(3):623-39. doi: 10.1084/jem.20121610. Epub 2013 Feb 25.
Results Reference
background
PubMed Identifier
18757300
Citation
Chabanon A, Desterke C, Rodenburger E, Clay D, Guerton B, Boutin L, Bennaceur-Griscelli A, Pierre-Louis O, Uzan G, Abecassis L, Bourgeade MF, Lataillade JJ, Le Bousse-Kerdiles MC. A cross-talk between stromal cell-derived factor-1 and transforming growth factor-beta controls the quiescence/cycling switch of CD34(+) progenitors through FoxO3 and mammalian target of rapamycin. Stem Cells. 2008 Dec;26(12):3150-61. doi: 10.1634/stemcells.2008-0219. Epub 2008 Aug 28.
Results Reference
background
PubMed Identifier
10989189
Citation
Zermati Y, Fichelson S, Valensi F, Freyssinier JM, Rouyer-Fessard P, Cramer E, Guichard J, Varet B, Hermine O. Transforming growth factor inhibits erythropoiesis by blocking proliferation and accelerating differentiation of erythroid progenitors. Exp Hematol. 2000 Aug;28(8):885-94. doi: 10.1016/s0301-472x(00)00488-4.
Results Reference
background
PubMed Identifier
18474728
Citation
Zhou L, Nguyen AN, Sohal D, Ying Ma J, Pahanish P, Gundabolu K, Hayman J, Chubak A, Mo Y, Bhagat TD, Das B, Kapoun AM, Navas TA, Parmar S, Kambhampati S, Pellagatti A, Braunchweig I, Zhang Y, Wickrema A, Medicherla S, Boultwood J, Platanias LC, Higgins LS, List AF, Bitzer M, Verma A. Inhibition of the TGF-beta receptor I kinase promotes hematopoiesis in MDS. Blood. 2008 Oct 15;112(8):3434-43. doi: 10.1182/blood-2008-02-139824. Epub 2008 May 12.
Results Reference
background
PubMed Identifier
29331635
Citation
Komrokji R, Garcia-Manero G, Ades L, Prebet T, Steensma DP, Jurcic JG, Sekeres MA, Berdeja J, Savona MR, Beyne-Rauzy O, Stamatoullas A, DeZern AE, Delaunay J, Borthakur G, Rifkin R, Boyd TE, Laadem A, Vo B, Zhang J, Puccio-Pick M, Attie KM, Fenaux P, List AF. Sotatercept with long-term extension for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes: a phase 2, dose-ranging trial. Lancet Haematol. 2018 Feb;5(2):e63-e72. doi: 10.1016/S2352-3026(18)30002-4. Epub 2018 Jan 10.
Results Reference
background
PubMed Identifier
28870615
Citation
Platzbecker U, Germing U, Gotze KS, Kiewe P, Mayer K, Chromik J, Radsak M, Wolff T, Zhang X, Laadem A, Sherman ML, Attie KM, Giagounidis A. Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol. 2017 Oct;18(10):1338-1347. doi: 10.1016/S1470-2045(17)30615-0. Epub 2017 Sep 1. Erratum In: Lancet Oncol. 2017 Oct;18(10):e562.
Results Reference
background
PubMed Identifier
27053300
Citation
Zhang H, Kozono DE, O'Connor KW, Vidal-Cardenas S, Rousseau A, Hamilton A, Moreau L, Gaudiano EF, Greenberger J, Bagby G, Soulier J, Grompe M, Parmar K, D'Andrea AD. TGF-beta Inhibition Rescues Hematopoietic Stem Cell Defects and Bone Marrow Failure in Fanconi Anemia. Cell Stem Cell. 2016 May 5;18(5):668-81. doi: 10.1016/j.stem.2016.03.002. Epub 2016 Mar 24.
Results Reference
background
PubMed Identifier
26309397
Citation
Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA, Slapak CA, Lahn MM. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther. 2015 Aug 10;9:4479-99. doi: 10.2147/DDDT.S86621. eCollection 2015.
Results Reference
background
PubMed Identifier
23403314
Citation
Badalucco S, Di Buduo CA, Campanelli R, Pallotta I, Catarsi P, Rosti V, Kaplan DL, Barosi G, Massa M, Balduini A. Involvement of TGFbeta1 in autocrine regulation of proplatelet formation in healthy subjects and patients with primary myelofibrosis. Haematologica. 2013 Apr;98(4):514-7. doi: 10.3324/haematol.2012.076752. Epub 2013 Feb 12.
Results Reference
background
PubMed Identifier
17473062
Citation
Ciurea SO, Merchant D, Mahmud N, Ishii T, Zhao Y, Hu W, Bruno E, Barosi G, Xu M, Hoffman R. Pivotal contributions of megakaryocytes to the biology of idiopathic myelofibrosis. Blood. 2007 Aug 1;110(3):986-93. doi: 10.1182/blood-2006-12-064626. Epub 2007 May 1.
Results Reference
background
PubMed Identifier
17198875
Citation
Gastinne T, Vigant F, Lavenu-Bombled C, Wagner-Ballon O, Tulliez M, Chagraoui H, Villeval JL, Lacout C, Perricaudet M, Vainchenker W, Benihoud K, Giraudier S. Adenoviral-mediated TGF-beta1 inhibition in a mouse model of myelofibrosis inhibit bone marrow fibrosis development. Exp Hematol. 2007 Jan;35(1):64-74. doi: 10.1016/j.exphem.2006.08.016.
Results Reference
background
PubMed Identifier
7803262
Citation
Martyre MC, Romquin N, Le Bousse-Kerdiles MC, Chevillard S, Benyahia B, Dupriez B, Demory JL, Bauters F. Transforming growth factor-beta and megakaryocytes in the pathogenesis of idiopathic myelofibrosis. Br J Haematol. 1994 Sep;88(1):9-16. doi: 10.1111/j.1365-2141.1994.tb04970.x.
Results Reference
background
PubMed Identifier
23462118
Citation
Zingariello M, Martelli F, Ciaffoni F, Masiello F, Ghinassi B, D'Amore E, Massa M, Barosi G, Sancillo L, Li X, Goldberg JD, Rana RA, Migliaccio AR. Characterization of the TGF-beta1 signaling abnormalities in the Gata1low mouse model of myelofibrosis. Blood. 2013 Apr 25;121(17):3345-63. doi: 10.1182/blood-2012-06-439661. Epub 2013 Mar 5.
Results Reference
background
PubMed Identifier
22200991
Citation
Ponce CC, de Lourdes F Chauffaille M, Ihara SS, Silva MR. The relationship of the active and latent forms of TGF-beta1 with marrow fibrosis in essential thrombocythemia and primary myelofibrosis. Med Oncol. 2012 Dec;29(4):2337-44. doi: 10.1007/s12032-011-0144-1. Epub 2011 Dec 27.
Results Reference
background
PubMed Identifier
15726648
Citation
Bock O, Loch G, Schade U, von Wasielewski R, Schlue J, Kreipe H. Aberrant expression of transforming growth factor beta-1 (TGF beta-1) per se does not discriminate fibrotic from non-fibrotic chronic myeloproliferative disorders. J Pathol. 2005 Apr;205(5):548-57. doi: 10.1002/path.1744.
Results Reference
background
PubMed Identifier
14991065
Citation
Shehata M, Schwarzmeier JD, Hilgarth M, Hubmann R, Duechler M, Gisslinger H. TGF-beta1 induces bone marrow reticulin fibrosis in hairy cell leukemia. J Clin Invest. 2004 Mar;113(5):676-85. doi: 10.1172/JCI19540.
Results Reference
background
PubMed Identifier
27592389
Citation
Ceglia I, Dueck AC, Masiello F, Martelli F, He W, Federici G, Petricoin EF 3rd, Zeuner A, Iancu-Rubin C, Weinberg R, Hoffman R, Mascarenhas J, Migliaccio AR. Preclinical rationale for TGF-beta inhibition as a therapeutic target for the treatment of myelofibrosis. Exp Hematol. 2016 Dec;44(12):1138-1155.e4. doi: 10.1016/j.exphem.2016.08.007. Epub 2016 Aug 31.
Results Reference
background
PubMed Identifier
27810993
Citation
Margolskee E, Krichevsky S, Orazi A, Silver RT. Evaluation of bone marrow morphology is essential for assessing disease status in recombinant interferon alpha-treated polycythemia vera patients. Haematologica. 2017 Mar;102(3):e97-e99. doi: 10.3324/haematol.2016.153973. Epub 2016 Nov 3. No abstract available.
Results Reference
background
PubMed Identifier
21562050
Citation
Anand S, Stedham F, Beer P, Gudgin E, Ortmann CA, Bench A, Erber W, Green AR, Huntly BJ. Effects of the JAK2 mutation on the hematopoietic stem and progenitor compartment in human myeloproliferative neoplasms. Blood. 2011 Jul 7;118(1):177-81. doi: 10.1182/blood-2010-12-327593. Epub 2011 May 11.
Results Reference
background
PubMed Identifier
27069254
Citation
Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016 May 19;127(20):2391-405. doi: 10.1182/blood-2016-03-643544. Epub 2016 Apr 11.
Results Reference
background
PubMed Identifier
21536863
Citation
Scherber R, Dueck AC, Johansson P, Barbui T, Barosi G, Vannucchi AM, Passamonti F, Andreasson B, Ferarri ML, Rambaldi A, Samuelsson J, Birgegard G, Tefferi A, Harrison CN, Radia D, Mesa RA. The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): international prospective validation and reliability trial in 402 patients. Blood. 2011 Jul 14;118(2):401-8. doi: 10.1182/blood-2011-01-328955. Epub 2011 May 2.
Results Reference
background
PubMed Identifier
29522138
Citation
Mascarenhas J, Hoffman R, Talpaz M, Gerds AT, Stein B, Gupta V, Szoke A, Drummond M, Pristupa A, Granston T, Daly R, Al-Fayoumi S, Callahan JA, Singer JW, Gotlib J, Jamieson C, Harrison C, Mesa R, Verstovsek S. Pacritinib vs Best Available Therapy, Including Ruxolitinib, in Patients With Myelofibrosis: A Randomized Clinical Trial. JAMA Oncol. 2018 May 1;4(5):652-659. doi: 10.1001/jamaoncol.2017.5818.
Results Reference
background
PubMed Identifier
22375970
Citation
Harrison C, Kiladjian JJ, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, McQuitty M, Hunter DS, Levy R, Knoops L, Cervantes F, Vannucchi AM, Barbui T, Barosi G. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012 Mar 1;366(9):787-98. doi: 10.1056/NEJMoa1110556.
Results Reference
background

Learn more about this trial

Intra-patient Dose Escalation Study to Investigate Safety and Feasibility of Vactosertib in Treating Anemic MPN Patients

We'll reach out to this number within 24 hrs