Apoptotic Cells for Therapeutic Use in Cytokine Storm Associated With Sepsis- A Phase Ib Clinical Trial

Peter Vernon van Heerden, Avraham Abutbul, Sigal Sviri, Eitan Zlotnick, Ahmad Nama, Sebastian Zimro, Raja El-Amore, Yehudit Shabat, Barak Reicher, Batla Falah, Dror Mevorach, Peter Vernon van Heerden, Avraham Abutbul, Sigal Sviri, Eitan Zlotnick, Ahmad Nama, Sebastian Zimro, Raja El-Amore, Yehudit Shabat, Barak Reicher, Batla Falah, Dror Mevorach

Abstract

Background: Sepsis has no proven specific pharmacologic treatment and reported mortality ranges from 30%-45%. The primary aim of this phase IB study was to determine the safety profile of Allocetra™-OTS (early apoptotic cell) infusion in subjects presenting to the emergency room with sepsis. The secondary aims were to measure organ dysfunction, intensive care unit (ICU) and hospital stays, and mortality. Exploratory endpoints included measuring immune modulator agents to elucidate the mechanism of action.

Methods: Ten patients presenting to the emergency room at the Hadassah Medical Center with sepsis were enrolled in this phase Ib clinical study. Enrolled patients were males and females aged 51-83 years, who had a Sequential Organ Failure Assessment (SOFA) score ≥2 above baseline and were septic due to presumed infection. Allocetra™-OTS was administered as a single dose (day +1) or in two doses of 140×106 cells/kg on (day +1 and +3), following initiation of standard-of-care (SOC) treatment for septic patients. Safety was evaluated by serious adverse events (SAEs) and adverse events (AEs). Organ dysfunction, ICU and hospital stays, and mortality, were compared to historical controls. Immune modulator agents were measured using Luminex® multiplex analysis.

Results: All 10 patients had mild-to-moderate sepsis with SOFA scores ranging from 2-6 upon entering the study. No SAEs and no related AEs were reported. All 10 study subjects survived, while matched historical controls had a mortality rate of 27%. The study subjects exhibited rapid resolution of organ dysfunction and had significantly shorter ICU stays compared to matched historical controls (p<0.0001). All patients had both elevated pro- and anti-inflammatory cytokines, chemokines, and additional immune modulators that gradually decreased following treatment.

Conclusion: Administration of apoptotic cells to patients with mild-to-moderate sepsis was safe and had a significant immuno-modulating effect, leading to early resolution of the cytokine storm.

Clinical trial registration: ClinicalTrials.gov Identifier: NCT03925857. (https://ichgcp.net/clinical-trials-registry/NCT03925857).

Keywords: Inflammation; apoptotic cells; cell therapeutics; cytokine storm; pneumonia; sepsis.

Conflict of interest statement

PvH received honoraria from Enlivex Ltd as a consultant. DM is the founder, and CMO of Enlivex Therapeutics Ltd. YS and BR are part of the research team of Enlivex Therapeutics Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

Copyright © 2021 van Heerden, Abutbul, Sviri, Zlotnick, Nama, Zimro, el-Amore, Shabat, Reicher, Falah and Mevorach.

Figures

Figure 1
Figure 1
Acute phase markers. Complete blood count (CBC) and C-reactive protein (CRP). Blood counts of fresh peripheral patient blood ethylenediamine tetraacetic acid (EDTA) samples were performed using blood analyzer. The differential of white blood cells (WBC), i.e. WBC subpopulations, was calculated. ‘Screening’ is a time point with a blood count that was performed 24 ± 6h before treatment with Allocetra-OTS, to test for the patient’s eligibility for Allocetra-OTS treatment. ‘Day 1’ is a blood sample just prior to Allocetra-OTS infusion. Numbers (109 cells/L) of (A) WBCs, (B) neutrophils, and (C) lymphocytes, are shown. (D) CRP levels of all patients in the indicated time points. *Patients 09–12 received two doses of Allocetra-OTS (days 1 and 3).
Figure 2
Figure 2
Kaplan-Meier survival curve. A comparison of survival between the study group (n=10) and matched-historical controls from the same hospital (n=37) for (A) all patients, and (B) patients with pneumonia (n=5 for the study group, n=19 for the historical controls), is shown. Matched controls were selected from all patients admitted to ICU with a diagnosis of sepsis between 2014–2019 and were matched by gender, age ± 7, Sequential Organ Failure Assessment (SOFA) score ±2 at presentation, and source of infection.
Figure 3
Figure 3
SOFA score progression during sepsis. (A) SOFA score at presentation compared to maximal SOFA score. (B–D) Change in SOFA score before administration of Allocetra-OTS and day 5 (B), day 7 (C), and day 28 (D). Average delta area under the curve (AUC) in the first (E) five days, and (F) seven days. Matched controls were selected from all patients admitted to ICU under the diagnosis of sepsis between 2014–2019 and were matched by gender, age ±7 years, SOFA score ±2 at presentation, and source of infection. Data is presented as the median within the interquartile range (IQR); mean values are marked with a ‘+’ sign; error bars represent the 10-90 percentile, with outliers presented.
Figure 4
Figure 4
Hospital and ICU/IMU duration of stay. The duration of stay for all patients in the study and for all patients in the matched historical control group who survived is presented (A) in the hospital and (B) in the Intensive Care Unit (ICU) or the Intermediate Medical Unit (IMU). Kaplan-Meier curves describing time-to-discharge from the (C) hospital and (D) ICU/IMU are presented. Matched controls were selected from all patients admitted to ICU/IMU with the diagnosis of sepsis between 2014–2019 and were matched by gender, age ±7, SOFA score ±2 at presentation, and source of infection. Data is presented as the median within the interquartile range (IQR); mean values are marked with a ‘+’ sign; error bars represent the 10–90 percentile, with outliers presented.
Figure 5
Figure 5
Pro-inflammatory cytokine kinetics during sepsis. Serum was obtained from patients at the indicated times and cytokine analysis was performed as described in Methods. The serum of three healthy volunteers was analyzed using the same methods and values are presented as the normal range (median ± range). Serum concentrations of (A) IL-6, (B) TNF-α, (C) IL-1β, (D) IL-18, and (E) IFN-γ are presented. Patients 01–08 received one dose of Allocetra-OTS on day 1 and patients 09–12 received two doses on days 1 and 3.
Figure 6
Figure 6
Anti-Inflammatory cytokine and growth factor kinetics during sepsis. Serum was obtained from patients at the indicated times and cytokine/growth factor analysis was performed as described in Methods. The serum of three healthy volunteers was analyzed using the same methods and values are presented as the normal range (median ± range). Serum concentrations of (A) IL-10, (B) IL-1Ra, (C) TNFR-1 are presented in the upper panel, (D) G-CSF, (E) VEGF, and (F) GM-CSF are presented in the lower panel. Patients 01–08 received one dose of Allocetra-OTS on day 1 and patients 09–12 received two doses on days 1 and 3.
Figure 7
Figure 7
Chemokine kinetics during sepsis. Serum was obtained from patients at the indicated times and cytokine/growth factor analysis was performed as described in Methods. The serum of three healthy volunteers was analyzed using the same methods and values are presented as the normal range (median ± range). Serum concentrations of (A) MCP-1, (B) IP-10, (C) MIP-1α, (D) IL-8, (E) GRO-β, and (F) RANTES are presented. Patients 01–08 received one dose of Allocetra-OTS on day 1 and patients 09–12 received two doses on days 1 and 3.
Figure 8
Figure 8
Immune modulator and endocrine hormone kinetics during sepsis. Serum was obtained from patients at the indicated times and cytokine/growth factors analysis was performed as described in Methods. The serum of three healthy volunteers was analyzed using the same methods and values presented as the normal range (median ± range). Serum concentrations of (A) TREM-1, (B) osteopontin, (C) N-GAL, (D) ghrelin, (E) leptin, and (F) glucagon, (G) cortisol, and (H) FT3 are presented in the lower panel. Patients 01–08 received one dose of Allocetra-OTS on day 1 and patients 09–12 received two doses on days 1 and 3.
Figure 9
Figure 9
Correlation matrix of cytokines, chemokines, and immunomodulators with SOFA score, CRP, and blood counts. A correlation heat-map of the tested cytokines/chemokines, HGFs, and immunomodulators with CRP, SOFA score, and blood counts. Pearson’s r values are on the top right and the Spearman’s ρ values are on the bottom left.

References

    1. Martin-Loeches I, Levy MM, Artigas A. Management of Severe Sepsis: Advances, Challenges, and Current Status. Drug Des Devel Ther (2015) 9:2079–88. doi: 10.2147/DDDT.S78757
    1. Rello J, Valenzuela-Sanchez F, Ruiz-Rodriguez M, Moyano S. Sepsis: A Review of Advances in Management. Adv Ther (2017) 34(11):2393–411. doi: 10.1007/s12325-017-0622-8
    1. Venkatesh B, Finfer S, Cohen J, Rajbhandari D, Arabi Y, Bellomo R, et al. . Adjunctive Glucocorticoid Therapy in Patients With Septic Shock. N Engl J Med (2018) 378(9):797–808. doi: 10.1056/NEJMoa1705835
    1. Vincent JL, Grimaldi D. Novel Interventions: What’s New and the Future. Crit Care Clin (2018) 34(1):161–73. doi: 10.1016/j.ccc.2017.08.012
    1. Finfer S, Bellomo R, Lipman J, French C, Dobb G, Myburgh J. Adult-Population Incidence of Severe Sepsis in Australian and New Zealand Intensive Care Units. Intensive Care Med (2004) 30(4):589–96. doi: 10.1007/s00134-004-2157-0
    1. Fleischmann C, Scherag A, Adhikari NK, Hartog CS, Tsaganos T, Schlattmann P, et al. . Assessment of Global Incidence and Mortality of Hospital-Treated Depsis. Current Estimates and Limitations. Am J Respir Crit Care Med (2016) 193(3):259–72. doi: 10.1164/rccm.201504-0781OC
    1. Liu V, Escobar GJ, Greene JD, Soule J, Whippy A, Angus DC, et al. . Hospital Deaths in Patients With Sepsis From 2 Independent Cohorts. JAMA (2014) 312(1):90–2. doi: 10.1001/jama.2014.5804
    1. Machado FR, Cavalcanti AB, Bozza FA, Ferreira EM, Angotti Carrara FS, Sousa JL, et al. . The Epidemiology of Sepsis in Brazilian Intensive Care Units (the Sepsis Prevalence Assessment Database, SPREAD): An Observational Study. Lancet Infect Dis (2017) 17(11):1180–9. doi: 10.1016/S1473-3099(17)30322-5
    1. Reinhart K, Daniels R, Kissoon N, Machado FR, Schachter RD, Finfer S. Recognizing Sepsis as a Global Health Priority — a WHO Resolution. N Engl J Med (2017) 377(5):414–7. doi: 10.1056/NEJMp1707170
    1. Rhee C, Dantes R, Epstein L, Murphy DJ, Seymour CW, Iwashyna TJ, et al. . Incidence and Trends of Sepsis in US Hospitals Using Clinical vs Claims Data, 2009-2014. JAMA (2017) 318(13):1241–9. doi: 10.1001/jama.2017.13836
    1. Hotchkiss RS, Moldawer LL, Opal SM, Reinhart K, Turnbull IR, Vincent JL. Sepsis and Septic Shock. Nat Rev Dis Primers (2016) 2:16045. doi: 10.1038/nrdp.2016.45
    1. Tang D, Kang R, Coyne CB, Zeh HJ, Lotze MT. Pamps and Damps: Signal 0s That Spur Autophagy and Immunity. Immunol Rev (2012) 249(1):158–75. doi: 10.1111/j.1600-065X.2012.01146.x
    1. Trahtemberg U, Mevorach D. Apoptotic Cells Induced Signaling for Immune Homeostasis in Macrophages and Dendritic Cells. Front Immunol (2017) 8:1356. doi: 10.3389/fimmu.2017.01356
    1. Mevorach D, Zuckerman T, Reiner I, Shimoni A, Samuel S, Nagler A, et al. . Single Infusion of Donor Mononuclear Early Apoptotic Cells as Prophylaxis for Graft-Versus-Host Disease in Myeloablative HLA-Matched Allogeneic Bone Marrow Transplantation: A Phase I/Iia Clinical Trial. Biol Blood Marrow Transplant (2014) 20(1):58–65. doi: 10.1016/j.bbmt.2013.10.010
    1. Rowan KM, Kerr JH, Major E, McPherson K, Short A, Vessey MP. Intensive Care Society’s APACHE II Study in Britain and Ireland–I: Variations in Case Mix of Adult Admissions to General Intensive Care Units and Impact on Outcome. BMJ (1993) 307(6910):972–7. doi: 10.1136/bmj.307.6910.972
    1. Knaus WA, Draper EA, Wagner DP, Zimmerman JE. APACHE II: A Severity of Disease Classification System. Crit Care Med (1985) 13(10):818–29. doi: 10.1097/00003246-198510000-00009
    1. Minne L, Abu-Hanna A, de Jonge E. Evaluation of SOFA-Based Models for Predicting Mortality in the ICU: A Systematic Review. Crit Care (2008) 12(6):R161. doi: 10.1186/cc7160
    1. Moon BH, Park SK, Jang DK, Jang KS, Kim JT, Han YM. Use of APACHE II and SAPS II to Predict Mortality for Hemorrhagic and Ischemic Stroke Patients. J Clin Neurosci (2015) 22(1):111–5. doi: 10.1016/j.jocn.2014.05.031
    1. Vincent JL, Moreno R. Clinical Review: Scoring Systems in the Critically Ill. Crit Care (2010) 14(2):207. doi: 10.1186/cc8204
    1. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. . The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA (2016) 315(8):801–10. doi: 10.1001/jama.2016.0287
    1. Raith EP, Udy AA, Bailey M, McGloughlin S, MacIsaac C, Bellomo R, et al. . Prognostic Accuracy of the SOFA Score, SIRS Criteria, and Qsofa Score for in-Hospital Mortality Among Adults With Suspected Infection Admitted to the Intensive Care Unit. JAMA (2017) 317(3):290–300. doi: 10.1001/jama.2016.20328
    1. Vincent JL, Moreno R, Takala J, Willatts S, De Mendonca A, Bruining H, et al. . The SOFA (Sepsis-Related Organ Failure Assessment) Score to Describe Organ Dysfunction/Failure. On Behalf of the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine. Intensive Care Med (1996) 22(7):707–10. doi: 10.1007/BF01709751
    1. Chaudhry H, Zhou J, Zhong Y, Ali MM, McGuire F, Nagarkatti PS, et al. . Role of Cytokines as a Double-Edged Sword in Sepsis. In Vivo (2013) 27(6):669–84.
    1. Chousterman BG, Swirski FK, Weber GF. Cytokine Storm and Sepsis Disease Pathogenesis. Semin Immunopathol (2017) 39(5):517–28. doi: 10.1007/s00281-017-0639-8
    1. Chousterman BG, Arnaud M. Is There a Role for Hematopoietic Growth Factors During Sepsis? Front Immunol (2018) 9:1015. doi: 10.3389/fimmu.2018.01015
    1. Aziz M, Jacob A, Yang WL, Matsuda A, Wang P. Current Trends in Inflammatory and Immunomodulatory Mediators in Sepsis. J Leukoc Biol (2013) 93(3):329–42. doi: 10.1189/jlb.0912437
    1. Jones BJ, Tan T, Bloom SR. Minireview: Glucagon in Stress and Energy Homeostasis. Endocrinology (2012) 153(3):1049–54. doi: 10.1210/en.2011-1979
    1. Meyer S, Schuetz P, Wieland M, Nusbaumer C, Mueller B, Christ-Crain M. Low Triiodothyronine Syndrome: A Prognostic Marker for Outcome in Sepsis? Endocrine (2011) 39(2):167–74. doi: 10.1007/s12020-010-9431-4
    1. Plikat K, Langgartner J, Buettner R, Bollheimer LC, Woenckhaus U, Scholmerich J, et al. . Frequency and Outcome of Patients With Nonthyroidal Illness Syndrome in a Medical Intensive Care Unit. Metabolism (2007) 56(2):239–44. doi: 10.1016/j.metabol.2006.09.020
    1. Wiersinga WJ, Seymour CW. Handbook of Sepsis. Cham, Switzerland: Springer Int. Publ. AG; (2018).
    1. Bonnefoy F, Gauthier T, Vallion R, Martin-Rodriguez O, Missey A, Daoui A, et al. . Factors Produced by Macrophages Eliminating Apoptotic Cells Demonstrate Pro-Resolutive Properties and Terminate Ongoing Inflammation. Front Immunol (2018) 9:2586. doi: 10.3389/fimmu.2018.02586
    1. Poon IK, Lucas CD, Rossi AG, Ravichandran KS. Apoptotic Cell Clearance: Basic Biology and Therapeutic Potential. Nat Rev Immunol (2014) 14(3):166–80. doi: 10.1038/nri3607
    1. Saas P, Daguindau E, Perruche S. Concise Review: Apoptotic Cell-Based Therapies-Rationale, Preclinical Results and Future Clinical Developments. Stem Cells (2016) 34(6):1464–73. doi: 10.1002/stem.2361
    1. Krispin A, Bledi Y, Atallah M, Trahtemberg U, Verbovetski I, Nahari E, et al. . Apoptotic Cell Thrombospondin-1 and Heparin-Binding Domain Lead to Dendritic-Cell Phagocytic and Tolerizing States. Blood (2006) 108(10):3580–9. doi: 10.1182/blood-2006-03-013334
    1. Beer L, Mildner M, Gyöngyösi M, Ankersmit HJ. Peripheral Blood Mononuclear Cell Secretome for Tissue Repair. Apoptosis (2016) 21(12):1336–53. doi: 10.1007/s10495-016-1292-8
    1. Hoetzenecker K, Assinger A, Lichtenauer M, Mildner M, Schweiger T, Starlinger P, et al. . Secretome of Apoptotic Peripheral Blood Cells (APOSEC) Attenuates Microvascular Obstruction in a Porcine Closed Chest Reperfused Acute Myocardial Infarction Model: Role of Platelet Aggregation and Vasodilation. Basic Res Cardiol (2012) 107(5):292–2. doi: 10.1007/s00395-012-0292-2
    1. Lichtenauer M, Mildner M, Baumgartner A, Hasun M, Werba G, Beer L, et al. . Intravenous and Intramyocardial Injection of Apoptotic White Blood Cell Suspensions Prevents Ventricular Remodelling by Increasing Elastin Expression in Cardiac Scar Tissue After Myocardial Infarction. Basic Res Cardiol (2011) 106(4):645–55. doi: 10.1007/s00395-011-0173-0
    1. Lichtenauer M, Mildner M, Hoetzenecker K, Zimmermann M, Podesser BK, Sipos W, et al. . Secretome of Apoptotic Peripheral Blood Cells (APOSEC) Confers Cytoprotection to Cardiomyocytes and Inhibits Tissue Remodelling After Acute Myocardial Infarction: A Preclinical Study. Basic Res Cardiol (2011) 106(6):1283–97. doi: 10.1007/s00395-011-0224-6
    1. Hacker S, Mittermayr R, Nickl S, Haider T, Lebherz-Eichinger D, Beer L, et al. . Paracrine Factors From Irradiated Peripheral Blood Mononuclear Cells Improve Skin Regeneration and Angiogenesis in a Porcine Burn Model. Sci Rep (2016) 6(1):25168. doi: 10.1038/srep25168
    1. Lee YJ, Moon C, Lee SH, Park HJ, Seoh JY, Cho MS, et al. . Apoptotic Cell Instillation After Bleomycin Attenuates Lung Injury Through Hepatocyte Growth Factor Induction. Eur Respir J (2012) 40(2):424–35. doi: 10.1183/09031936.00096711
    1. Ren Y, Xie Y, Jiang G, Fan J, Yeung J, Li W, et al. . Apoptotic Cells Protect Mice Against Lipopolysaccharide-Induced Shock. J Immunol (2008) 180(7):4978–85. doi: 10.4049/jimmunol.180.7.4978
    1. Kasiri MM, Beer L, Nemec L, Gruber F, Pietkiewicz S, Haider T, et al. . Dying Blood Mononuclear Cell Secretome Exerts Antimicrobial Activity. Eur J Clin Invest (2016) 46:853–63. doi: 10.1111/eci.12667
    1. Cohen J, Opal S, Calandra T. Sepsis Studies Need New Direction. Lancet Infect Dis (2012) 12(7):503–5. doi: 10.1016/S1473-3099(12)70136-6
    1. Tamayo E, Fernandez A, Almansa R, Carrasco E, Heredia M, Lajo C, et al. . Pro- and Anti-Inflammatory Responses Are Regulated Simultaneously From the First Moments of Septic Shock. Eur Cytokine Netw (2011) 22(2):82–7. doi: 10.1684/ecn.2011.0281
    1. Li X, Xu Z, Pang X, Huang Y, Yang B, Yang Y, et al. . Interleukin-10/Lymphocyte Ratio Predicts Mortality in Severe Septic Patients. PloS One (2017) 12(6):e0179050. doi: 10.1371/journal.pone.0179050
    1. Libraty DH, Endy TP, Houng HS, Green S, Kalayanarooj S, Suntayakorn S, et al. . Differing Influences of Virus Burden and Immune Activation on Disease Severity in Secondary Dengue-3 Virus Infections. J Infect Dis (2002) 185(9):1213–21. doi: 10.1086/340365
    1. Pugin J, Ricou B, Steinberg KP, Suter PM, Martin TR. Proinflammatory Activity in Bronchoalveolar Lavage Fluids From Patients With ARDS, a Prominent Role for Interleukin-1. Am J Respir Crit Care Med (1996) 153(6 Pt 1):1850–6. doi: 10.1164/ajrccm.153.6.8665045
    1. Gauthier J, Turtle CJ. Insights Into Cytokine Release Syndrome and Neurotoxicity After CD19-Specific CAR-T Cell Therapy. Curr Res Transl Med (2018) 66(2):50–2. doi: 10.1016/j.retram.2018.03.003
    1. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T Cell-Induced Cytokine Release Syndrome Is Mediated by Macrophages and Abated by IL-1 Blockade. Nat Med (2018) 24(6):731–8. doi: 10.1038/s41591-018-0041-7
    1. Huang R, Xia J, Chen Y, Shan C, Wu C. A Family Cluster of SARS-CoV-2 Infection Involving 11 Patients in Nanjing, China. Lancet Infect Dis (2020) 20(5):534–5. doi: 10.1016/S1473-3099(20)30147-X
    1. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ, et al. . COVID-19: Consider Cytokine Storm Syndromes and Immunosuppression. Lancet (2020) 395(10229):1033–4. doi: 10.1016/S0140-6736(20)30628-0

Source: PubMed

3
購読する