A phase I, first-in-human study to evaluate the safety and tolerability, pharmacokinetics, and pharmacodynamics of MRG-001 in healthy subjects

Ali R Ahmadi, George Atiee, Bart Chapman, Laurie Reynolds, John Sun, Andrew M Cameron, Russell N Wesson, James F Burdick, Zhaoli Sun, Ali R Ahmadi, George Atiee, Bart Chapman, Laurie Reynolds, John Sun, Andrew M Cameron, Russell N Wesson, James F Burdick, Zhaoli Sun

Abstract

Preclinical studies demonstrate that pharmacological mobilization and recruitment of endogenous bone marrow stem cells and immunoregulatory cells by a fixed-dose drug combination (MRG-001) improves wound healing, promotes tissue regeneration, and prevents allograft rejection. In this phase I, first-in-human study, three cohorts receive subcutaneous MRG-001 or placebo, every other day for 5 days. The primary outcome is safety and tolerability of MRG-001. Fourteen subjects received MRG-001 and seven received a placebo. MRG-001 is safe over the selected dose range. There are no clinically significant laboratory changes. The intermediate dose group demonstrates the most significant white blood cell, stem cell, and immunoregulatory cell mobilization. PBMC RNA sequencing and gene set enrichment analysis reveal 31 down-regulated pathways in the intermediate MRG-001 dose group compared with no changes in the placebo group. MRG-001 is safe across all dose ranges. MRG-001 may be a clinically useful therapy for immunoregulation and tissue regeneration (ClinicalTrials.gov: NCT04646603).

Keywords: MRG-001; clinical trial; healthy volunteers; pharmacodynamics; pharmacokinetics; phase I; plerixafor; safety; stem cells; tacrolimus.

Conflict of interest statement

Declaration of interests A.R.A. is a consultant to MedRegen LLC. J.B. and J.S. are employees of MedRegen LLC. J.B. and Z.S. are shareholders in MedRegen LLC. Z.S. is also the founder of MedRegen LLC.

Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
MRG-001 pharmacokinetics (A) Time profiles for plerixafor following SC administration of a single dose of 0.005, 0.01, and 0.02 mL/kg MRG-001 on day 1. (B) Time profiles for plerixafor following SC administration of 0.005, 0.01, and 0.02 mL/kg MRG-001 on day 5. (C) Time profiles for tacrolimus following SC administration of a single dose of 0.005, 0.01, and 0.02 mL/kg MRG-001 on day 1. (D) Time profiles for tacrolimus following SC administration of 0.005, 0.01, and 0.02 mL/kg MRG-001 on day 5. n = 4 per group. Bars represent standard deviation of the mean.
Figure 2
Figure 2
WBC mobilization with MRG-001 in healthy subjects Subjects received SC injections every other day of saline (placebo control, n = 6), low-dose (0.005 mL/kg), mid-dose (0.01 mL/kg), or high-dose (0.02 mL/kg) MRG-001 (n = 4/group). Venous blood was collected to determine the WBC differential count at several time intervals. (A) WBCs on day 1 after a single dose SC injection of saline or MRG-001. (B) WBCs on day 5 after the third dose SC injection of saline or MRG-001. Each value represents the mean ± SD. The gray area represents the normal reference range.
Figure 3
Figure 3
Lymphocyte mobilization with MRG-001 in healthy subjects Subjects received SC injections every other day of saline (placebo control), mid-dose (0.01 mL/kg), or high-dose (0.02 mL/kg) MRG-001 (n = 4/group). Venous blood was collected and PBMCs were isolated at different time intervals on day 1 after a single dose SC injection of saline or MRG-001 and on day 5 after the third dose. Lymphocyte populations (CD3+, CD3+CD4+, CD3+CD8+, CD19+, CD3+CD4+Foxp3+, and CD3+CD8+Foxp3+) in PBMC were analyzed by flow cytometry. At each blood draw timepoint, absolute number of circulating lymphocyte subsets, such as CD3+ T cells, was calculated by multiplication of CD3+ cell fraction of PBMC by the total circulating PBMC count. (A) Lymphocyte populations in peripheral blood on day 1 after a single dose SC injection of saline or MRG-001. (B) Lymphocyte populations in peripheral blood on day 5 after the third dose. Each value represents the mean ± SD. Yellow marked cells represent significant difference with p

Figure 4

Stem cell mobilization with MRG-001…

Figure 4

Stem cell mobilization with MRG-001 Subjects received SC injections every other day of…

Figure 4
Stem cell mobilization with MRG-001 Subjects received SC injections every other day of saline (placebo control), mid-dose (0.01 mL/kg), or high-dose (0.02 mL/kg) MRG-001 (n = 4/group). Venous blood was collected and PBMCs were isolated at different time intervals on day 1 after a single dose SC injection of saline or MRG-001 and on day 5 after the third dose. Stem cell populations (CD45IntCD34+, CD45IntCD34+CD90+, CD45IntCD34+CD133+, CD45IntCD34+CD133+CD31+, CD45IntCD34+CD133+VEGFR2+, CD45IntCD34+SSEA3+) in PBMC were analyzed by flow cytometry. At each blood draw timepoint, absolute number of circulating stem cells, such as CD45IntCD34+ HSCs, was calculated by multiplication of CD45IntCD34+ cell fraction of PBMC by the total circulating PBMC count. (A) Stem cells in peripheral blood on day 1 after a single dose SC injection of saline or MRG-001. (B) Stem cells in peripheral blood on day 5 after the third dose. Each value represents the mean ± SD. Yellow marked cells represent significant difference with p

Figure 5

Regulated molecular pathways by MRG-001…

Figure 5

Regulated molecular pathways by MRG-001 RNA sequencing transcriptome studies of PBMCs were performed…

Figure 5
Regulated molecular pathways by MRG-001 RNA sequencing transcriptome studies of PBMCs were performed at different time points following a single dose SC injection of saline (placebo) or MRG-001. (A) GSEA for interpreting gene expression profiles revealed 17, 24 and 19 down-regulated pathways at 8, 12, and 24 h after administration of 0.01 mL/kg MRG-001. A total of 31 down-regulated pathways were recognized and summarized in a Venn diagram. (B) GSEA showed 4, 9, 5, and 8 down-regulated pathways at 8, 12, 24, and 48 h, and 1 up-regulated pathway at 8, 24, and 48 h after administration of 0.02 mL/kg MRG-001. A total of 14 down-regulated and 2 up-regulated pathways were recognized, and down-regulated pathways were summarized in a Venn diagram.
Similar articles
Cited by
References
    1. Okabayashi T., Cameron A.M., Hisada M., Montgomery R.A., Williams G.M., Sun Z. Mobilization of host stem cells enables long-term liver transplant acceptance in a strongly rejecting rat strain combination. Am. J. Transplant. 2011;11:2046–2056. doi: 10.1111/j.1600-6143.2011.03698.x. - DOI - PMC - PubMed
    1. Lin Q., Wesson R.N., Maeda H., Wang Y., Cui Z., Liu J.O., Cameron A.M., Gao B., Montgomery R.A., Williams G.M., Sun Z. Pharmacological mobilization of endogenous stem cells significantly promotes skin regeneration after full-thickness excision: the synergistic activity of AMD3100 and tacrolimus. J. Invest. Dermatol. 2014;134:2458–2468. doi: 10.1038/jid.2014.162. - DOI - PMC - PubMed
    1. Brave M., Farrell A., Ching Lin S., Ocheltree T., Pope Miksinski S., Lee S.L., Saber H., Fourie J., Tornoe C., Booth B., et al. FDA review summary: Mozobil in combination with granulocyte colony-stimulating factor to mobilize hematopoietic stem cells to the peripheral blood for collection and subsequent autologous transplantation. Oncology. 2010;78:282–288. doi: 10.1159/000315736. - DOI - PubMed
    1. Hu X., Okabayashi T., Cameron A.M., Wang Y., Hisada M., Li J., Raccusen L.C., Zheng Q., Montgomery R.A., Williams G.M., Sun Z. Chimeric Allografts Induced by Short-Term Treatment With Stem Cell-Mobilizing Agents Result in Long-Term Kidney Transplant Survival Without Immunosuppression: A Study in Rats. Am. J. Transplant. 2016;16:2055–2065. doi: 10.1111/ajt.13706. - DOI - PMC - PubMed
    1. Cameron A.M., Wesson R.N., Ahmadi A.R., Singer A.L., Hu X., Okabayashi T., Wang Y., Shigoka M., Fu Y., Gao W., et al. Chimeric Allografts Induced by Short-Term Treatment With Stem Cell Mobilizing Agents Result in Long-Term Kidney Transplant Survival Without Immunosuppression: II, Study in Miniature Swine. Am. J. Transplant. 2016;16:2066–2076. doi: 10.1111/ajt.13703. - DOI - PubMed
Show all 35 references
Publication types
Associated data
Related information
[x]
Cite
Copy Download .nbib .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Stem cell mobilization with MRG-001 Subjects received SC injections every other day of saline (placebo control), mid-dose (0.01 mL/kg), or high-dose (0.02 mL/kg) MRG-001 (n = 4/group). Venous blood was collected and PBMCs were isolated at different time intervals on day 1 after a single dose SC injection of saline or MRG-001 and on day 5 after the third dose. Stem cell populations (CD45IntCD34+, CD45IntCD34+CD90+, CD45IntCD34+CD133+, CD45IntCD34+CD133+CD31+, CD45IntCD34+CD133+VEGFR2+, CD45IntCD34+SSEA3+) in PBMC were analyzed by flow cytometry. At each blood draw timepoint, absolute number of circulating stem cells, such as CD45IntCD34+ HSCs, was calculated by multiplication of CD45IntCD34+ cell fraction of PBMC by the total circulating PBMC count. (A) Stem cells in peripheral blood on day 1 after a single dose SC injection of saline or MRG-001. (B) Stem cells in peripheral blood on day 5 after the third dose. Each value represents the mean ± SD. Yellow marked cells represent significant difference with p

Figure 5

Regulated molecular pathways by MRG-001…

Figure 5

Regulated molecular pathways by MRG-001 RNA sequencing transcriptome studies of PBMCs were performed…

Figure 5
Regulated molecular pathways by MRG-001 RNA sequencing transcriptome studies of PBMCs were performed at different time points following a single dose SC injection of saline (placebo) or MRG-001. (A) GSEA for interpreting gene expression profiles revealed 17, 24 and 19 down-regulated pathways at 8, 12, and 24 h after administration of 0.01 mL/kg MRG-001. A total of 31 down-regulated pathways were recognized and summarized in a Venn diagram. (B) GSEA showed 4, 9, 5, and 8 down-regulated pathways at 8, 12, 24, and 48 h, and 1 up-regulated pathway at 8, 24, and 48 h after administration of 0.02 mL/kg MRG-001. A total of 14 down-regulated and 2 up-regulated pathways were recognized, and down-regulated pathways were summarized in a Venn diagram.
Figure 5
Figure 5
Regulated molecular pathways by MRG-001 RNA sequencing transcriptome studies of PBMCs were performed at different time points following a single dose SC injection of saline (placebo) or MRG-001. (A) GSEA for interpreting gene expression profiles revealed 17, 24 and 19 down-regulated pathways at 8, 12, and 24 h after administration of 0.01 mL/kg MRG-001. A total of 31 down-regulated pathways were recognized and summarized in a Venn diagram. (B) GSEA showed 4, 9, 5, and 8 down-regulated pathways at 8, 12, 24, and 48 h, and 1 up-regulated pathway at 8, 24, and 48 h after administration of 0.02 mL/kg MRG-001. A total of 14 down-regulated and 2 up-regulated pathways were recognized, and down-regulated pathways were summarized in a Venn diagram.

References

    1. Okabayashi T., Cameron A.M., Hisada M., Montgomery R.A., Williams G.M., Sun Z. Mobilization of host stem cells enables long-term liver transplant acceptance in a strongly rejecting rat strain combination. Am. J. Transplant. 2011;11:2046–2056. doi: 10.1111/j.1600-6143.2011.03698.x.
    1. Lin Q., Wesson R.N., Maeda H., Wang Y., Cui Z., Liu J.O., Cameron A.M., Gao B., Montgomery R.A., Williams G.M., Sun Z. Pharmacological mobilization of endogenous stem cells significantly promotes skin regeneration after full-thickness excision: the synergistic activity of AMD3100 and tacrolimus. J. Invest. Dermatol. 2014;134:2458–2468. doi: 10.1038/jid.2014.162.
    1. Brave M., Farrell A., Ching Lin S., Ocheltree T., Pope Miksinski S., Lee S.L., Saber H., Fourie J., Tornoe C., Booth B., et al. FDA review summary: Mozobil in combination with granulocyte colony-stimulating factor to mobilize hematopoietic stem cells to the peripheral blood for collection and subsequent autologous transplantation. Oncology. 2010;78:282–288. doi: 10.1159/000315736.
    1. Hu X., Okabayashi T., Cameron A.M., Wang Y., Hisada M., Li J., Raccusen L.C., Zheng Q., Montgomery R.A., Williams G.M., Sun Z. Chimeric Allografts Induced by Short-Term Treatment With Stem Cell-Mobilizing Agents Result in Long-Term Kidney Transplant Survival Without Immunosuppression: A Study in Rats. Am. J. Transplant. 2016;16:2055–2065. doi: 10.1111/ajt.13706.
    1. Cameron A.M., Wesson R.N., Ahmadi A.R., Singer A.L., Hu X., Okabayashi T., Wang Y., Shigoka M., Fu Y., Gao W., et al. Chimeric Allografts Induced by Short-Term Treatment With Stem Cell Mobilizing Agents Result in Long-Term Kidney Transplant Survival Without Immunosuppression: II, Study in Miniature Swine. Am. J. Transplant. 2016;16:2066–2076. doi: 10.1111/ajt.13703.
    1. Sun Z., Williams G.M. Host stem cells repopulate liver allografts: reverse chimerism. Chimerism. 2011;2:120–122. doi: 10.4161/chim.2.4.19177.
    1. Ray K. Transplantation: Host stem cells boost tolerance of liver grafts. Nat. Rev. Gastroenterol. Hepatol. 2011;8:601. doi: 10.1038/nrgastro.2011.178.
    1. Carney E.F. Transplantation: Reverse chimerism enables graft acceptance. Nat. Rev. Nephrol. 2016;12:125. doi: 10.1038/nrneph.2016.8.
    1. Tolar J., McGrath J.A. Augmentation of cutaneous wound healing by pharmacologic mobilization of endogenous bone marrow stem cells. J. Invest. Dermatol. 2014;134:2312–2314. doi: 10.1038/jid.2014.209.
    1. Qi L., Ahmadi A.R., Huang J., Chen M., Pan B., Kuwabara H., Iwasaki K., Wang W., Wesson R., Cameron A.M., et al. Major Improvement in Wound Healing Through Pharmacologic Mobilization of Stem Cells in Severely Diabetic Rats. Diabetes. 2020;69:699–712. doi: 10.2337/db19-0907.
    1. Zhai R., Wang Y., Qi L., Williams G.M., Gao B., Song G., Burdick J.F., Sun Z. Pharmacological Mobilization of Endogenous Bone Marrow Stem Cells Promotes Liver Regeneration after Extensive Liver Resection in Rats. Sci. Rep. 2018;8:3587. doi: 10.1038/s41598-018-21961-2.
    1. Iwasaki K., Ahmadi A.R., Qi L., Chen M., Wang W., Katsumata K., Tsuchida A., Burdick J., Cameron A.M., Sun Z. Pharmacological Mobilization and Recruitment of Stem Cells in Rats Stops Abdominal Adhesions After Laparotomy. Sci. Rep. 2019;9:7149. doi: 10.1038/s41598-019-43734-1.
    1. Li X., Sun Z. 2022. Methods of Treating Inflammatory Bowel Disease with AMD3100 and Tacrolimus.
    1. Peiffer B.J., Qi L., Ahmadi A.R., Wang Y., Guo Z., Peng H., Sun Z., Liu J.O. Activation of BMP Signaling by FKBP12 Ligands Synergizes with Inhibition of CXCR4 to Accelerate Wound Healing. Cell Chem. Biol. 2019;26:652–661.e4. doi: 10.1016/j.chembiol.2019.01.011.
    1. Notta F., Doulatov S., Laurenti E., Poeppl A., Jurisica I., Dick J.E. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science. 2011;333:218–221. doi: 10.1126/science.1201219.
    1. Lucey M.R., Terrault N., Ojo L., Hay J.E., Neuberger J., Blumberg E., Teperman L.W. Long-term management of the successful adult liver transplant: 2012 practice guideline by the American Association for the Study of Liver Diseases and the American Society of Transplantation. Liver Transpl. 2013;19:3–26. doi: 10.1002/lt.23566.
    1. Astellas Pharma US Inc . 2018. Prograft (Tacrolimus) Package Insert.
    1. Pantin J., Purev E., Tian X., Cook L., Donohue-Jerussi T., Cho E., Reger R., Hsieh M., Khuu H., Calandra G., et al. Effect of high-dose plerixafor on CD34(+) cell mobilization in healthy stem cell donors: results of a randomized crossover trial. Haematologica. 2017;102:600–609. doi: 10.3324/haematol.2016.147132.
    1. Wing J.B., Tanaka A., Sakaguchi S. Human FOXP3(+) Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity. 2019;50:302–316. doi: 10.1016/j.immuni.2019.01.020.
    1. Ni X., Wang Q., Gu J., Lu L. Clinical and Basic Research Progress on Treg-Induced Immune Tolerance in Liver Transplantation. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.535012.
    1. Muñoz-Rojas A.R., Mathis D. Tissue regulatory T cells: regulatory chameleons. Nat. Rev. Immunol. 2021;21:597–611. doi: 10.1038/s41577-021-00519-w.
    1. Rigato M., Bittante C., Albiero M., Avogaro A., Fadini G.P. Circulating Progenitor Cell Count Predicts Microvascular Outcomes in Type 2 Diabetic Patients. J. Clin. Endocrinol. Metab. 2015;100:2666–2672. doi: 10.1210/jc.2015-1687.
    1. Fadini G.P., Rigato M., Cappellari R., Bonora B.M., Avogaro A. Long-term Prediction of Cardiovascular Outcomes by Circulating CD34+ and CD34+CD133+ Stem Cells in Patients With Type 2 Diabetes. Diabetes Care. 2017;40:125–131. doi: 10.2337/dc16-1755.
    1. Hayek S.S., MacNamara J., Tahhan A.S., Awad M., Yadalam A., Ko Y.A., Healy S., Hesaroieh I., Ahmed H., Gray B., et al. Circulating Progenitor Cells Identify Peripheral Arterial Disease in Patients With Coronary Artery Disease. Circ. Res. 2016;119:564–571. doi: 10.1161/CIRCRESAHA.116.308802.
    1. Werner N., Kosiol S., Schiegl T., Ahlers P., Walenta K., Link A., Böhm M., Nickenig G. Circulating endothelial progenitor cells and cardiovascular outcomes. N. Engl. J. Med. 2005;353:999–1007. doi: 10.1056/NEJMoa043814.
    1. Rigato M., Avogaro A., Fadini G.P. Levels of Circulating Progenitor Cells, Cardiovascular Outcomes and Death: A Meta-Analysis of Prospective Observational Studies. Circ. Res. 2016;118:1930–1939. doi: 10.1161/CIRCRESAHA.116.308366.
    1. Patel R.S., Li Q., Ghasemzadeh N., Eapen D.J., Moss L.D., Janjua A.U., Manocha P., Kassem H.A., Veledar E., Samady H., et al. Circulating CD34+ progenitor cells and risk of mortality in a population with coronary artery disease. Circ. Res. 2015;116:289–297. doi: 10.1161/CIRCRESAHA.116.304187.
    1. Moazzami K., Lima B.B., Hammadah M., Ramadan R., Al Mheid I., Kim J.H., Alkhoder A., Obideen M., Levantsevych O., Shah A., et al. Association Between Change in Circulating Progenitor Cells During Exercise Stress and Risk of Adverse Cardiovascular Events in Patients With Coronary Artery Disease. JAMA Cardiol. 2020;5:147–155. doi: 10.1001/jamacardio.2019.4528.
    1. Fadini G.P., Schiavon M., Cantini M., Baesso I., Facco M., Miorin M., Tassinato M., de Kreutzenberg S.V., Avogaro A., Agostini C. Circulating progenitor cells are reduced in patients with severe lung disease. Stem Cell. 2006;24:1806–1813. doi: 10.1634/stemcells.2005-0440.
    1. Bonora B.M., Fogar P., Zuin J., Falaguasta D., Cappellari R., Cattelan A., Marinello S., Ferrari A., Avogaro A., Plebani M., et al. Hyperglycemia, Reduced Hematopoietic Stem Cells, and Outcome of COVID-19. Diabetes. 2022;71:788–794. doi: 10.2337/db21-0965.
    1. Muggeridge D., Dodd J., Ross M.D. CD34(+) progenitors are predictive of mortality and are associated with physical activity in cardiovascular disease patients. Atherosclerosis. 2021;333:108–115. doi: 10.1016/j.atherosclerosis.2021.07.004.
    1. Yang C.C., Sung P.H., Cheng B.C., Li Y.C., Chen Y.L., Lee M.S., Yip H.K. Safety and efficacy of intrarenal arterial autologous CD34+ cell transfusion in patients with chronic kidney disease: A randomized, open-label, controlled phase II clinical trial. Stem Cells Transl. Med. 2020;9:827–838. doi: 10.1002/sctm.19-0409.
    1. Salama H., Zekri A.R.N., Bahnassy A.A., Medhat E., Halim H.A., Ahmed O.S., Mohamed G., Al Alim S.A., Sherif G.M. Autologous CD34+ and CD133+ stem cells transplantation in patients with end stage liver disease. World J. Gastroenterol. 2010;16:5297–5305. doi: 10.3748/wjg.v16.i42.5297.
    1. Bonora B.M., Cappellari R., Mazzucato M., Rigato M., Grasso M., Menegolo M., Bruttocao A., Avogaro A., Fadini G.P. Stem cell mobilization with plerixafor and healing of diabetic ischemic wounds: A phase IIa, randomized, double-blind, placebo-controlled trial. Stem Cells Transl. Med. 2020;9:965–973. doi: 10.1002/sctm.20-0020.
    1. Duffy M.J., O'Grady S., Tang M., Crown J. MYC as a target for cancer treatment. Cancer Treat Rev. 2021;94 doi: 10.1016/j.ctrv.2021.102154.

Source: PubMed

3
Sottoscrivi