SBIR-STTR Award

Humanized Monoclonal Antibodies to Treat Mucormycosis
Award last edited on: 7/29/2020

Sponsored Program
SBIR
Awarding Agency
NIH : NIAID
Total Award Amount
$599,778
Award Phase
2
Solicitation Topic Code
-----

Principal Investigator
Gary Lazar

Company Information

Vitalex Biosciences LLC

2 Conifer
Irvine, CA 92620
   (323) 712-9380
   N/A
   N/A
Location: Single
Congr. District: 45
County: Orange

Phase I

Contract Number: 1R43AI138904-01A1
Start Date: 8/9/2018    Completed: 7/31/2020
Phase I year
2018
Phase I Amount
$300,000
Mucormycosis, most commonly caused by Rhizopus oryzae, is a life-threatening infection that occurs in patients immunocompromised by diabetic ketoacidosis (DKA), neutropenia, corticosteroid use, increased serum iron and/or severe trauma. Because of the rising prevalence of these risk factors, the incidence of mucormycosis has risen. Despite disfiguring surgery and aggressive antifungal therapy, the mortality of mucormycosis ranges from ~ 50% to 100%. The obvious unmet need for new, effective treatments and preventive strategies has been the driving force of our research program for over seventeen years. We propose to develop a passive vaccine targeting mucormycosis (i.e., an antibody that can be administered to patients with mucormycosis). Data in the academic laboratory of our founder (Dr. Ibrahim) indicate that antibody-based therapy is a promising strategy to treat mucormycosis. This technology is based on the important discovery that the fungal cell surface proteins encoded by CotH facilitate disease progression by allowing R. oryzae to invade mammalian cells via binding to Glucose Regulated Protein 78 (GRP78), a heat shock conserved protein expressed on endothelial cells lining blood vessels during mucormycosis. Importantly, CotH proteins were found to be conserved among Mucorales (organisms that cause mucormycosis) and absent from any other cell type including mammalian. Our data also show that CotH proteins are key determinants of mucormycosis pathogenesis since R. oryzae coth null mutants have markedly reduced virulence in mouse models of mucormycosis. Further, polyclonal antibodies targeting CotH are highly protective against murine mucormycosis caused by R. oryzae, Mucor, Lichtheimia, Cunninghamella, Rhizomucor, and Apophysomyces. Importantly and highly relevant to this application, anti-CotH murine monoclonal antibodies (mAb) raised against a peptide predicted to be present in the binding domain to GRP78, prevent the ability of R. oryzae to invade and injure endothelial cells in vitro and protect mice from mucormycosis caused by several Mucorales to levels that exceed those seen with antifungal therapy. While these mAbs are a promising new therapy for mucormycosis, the feasibility of further clinical development will hinge upon successful humanization of the Abs. Mouse mAbs cannot be used to treat humans, because humans mount an immune reaction to mouse mAbs that can cause rapid removal of the mAbs, systemic inflammation, severe allergic reactions, and even a risk for death. The humanization process prevents these undesirable effects. Thus, we propose two AIMS: 1) Develop our lead murine mAb into a humanized version with retained/enhanced binding ability to CotH proteins; and 2) Determine the protective activity of the humanized Ab in vitro/in vivo and evaluate its toxicity to human tissues. We propose conservative feasibility milestones that are part of a standard, methodical development pathway for our unique mAbs as a novel treatment for mucormycosis. The proposed work will identify a lead humanized Ab that will go into further development to ultimately test in clinical trials as an adjunctive therapy.

Project Terms:
Adrenal Cortex Hormones; Affinity; Allergic Reaction; Amino Acids; Angioinvasion; Animal Model; Antibiotics; Antibodies; Antibody Therapy; Antifungal Agents; Antifungal Therapy; Antigens; Attenuated; base; Binding; Biological Sciences; Blood Vessels; Brain; cancer transplantation; cell injury; Cell Line; Cell Surface Proteins; cell type; Cessation of life; cGMP production; Clinical; clinical development; Clinical Trials; clinically relevant; combat; cross reactivity; Cunninghamella; Data; Development; Diabetes Mellitus; Diabetic Ketoacidosis; Diagnostic; Disease; Disease Progression; Dose; driving force; effective therapy; Endothelial Cells; Enzyme-Linked Immunosorbent Assay; Erythrocytes; Excision; Fc Immunoglobulins; Feasibility Studies; Funding; Genes; glucose-regulated proteins; Goals; Grant; Heat shock proteins; Heat-Shock Response; Hematogenous; Hematopoietic Stem Cell Transplantation; Human; Human Cloning; human tissue; humanized monoclonal antibodies; IgG1; Immune system; Immunocompromised Host; immunoreaction; Immunotherapy; improved; In Vitro; in vitro activity; in vivo; Incidence; Infection; Inflammation; injured; Injury; Intervention; Invaded; Investments; Iron; Laboratories; Lead; lead candidate; Life; Mammalian Cell; Mediating; method development; Molds; Monoclonal Antibodies; mortality; mouse model; Mucor; Mucorales; Mucormycosis; murine monoclonal antibody; Mus; mutant; Mycoses; Necrosis; Neutropenia; novel; novel therapeutics; operation; Operative Surgical Procedures; Organ Transplantation; Organism; Orphan Drugs; Outcome; Pathogenesis; Pathway interactions; Patients; Penetration; Peptides; Phagocytosis; Pharmaceutical Preparations; Phase; polyclonal antibody; posaconazole; Positioning Attribute; pre-clinical; Prevalence; prevent; Prevention strategy; primary endpoint; Privatization; Process; programs; Proteins; Reporting; Research; Resources; Rhizomucor; Rhizopus; Risk; Risk Factors; secondary endpoint; Serum; Small Business Innovation Research Grant; Small Business Technology Transfer Research; Soldier; Stains; synergism; Technology; Testing; Therapeutics for Rare and Neglected Diseases; Thrombosis; Tissues; Toxic effect; Trauma; treatment strategy; Umbilical vein; United States National Institutes of Health; Vaccines; Virulence; Work; Zygomycosis;

Phase II

Contract Number: 5R43AI138904-02
Start Date: 8/9/2018    Completed: 7/31/2020
Phase II year
2019
Phase II Amount
$299,778
Mucormycosis, most commonly caused by Rhizopus oryzae, is a life-threatening infection that occurs in patients immunocompromised by diabetic ketoacidosis (DKA), neutropenia, corticosteroid use, increased serum iron and/or severe trauma. Because of the rising prevalence of these risk factors, the incidence of mucormycosis has risen. Despite disfiguring surgery and aggressive antifungal therapy, the mortality of mucormycosis ranges from ~ 50% to 100%. The obvious unmet need for new, effective treatments and preventive strategies has been the driving force of our research program for over seventeen years. We propose to develop a passive vaccine targeting mucormycosis (i.e., an antibody that can be administered to patients with mucormycosis). Data in the academic laboratory of our founder (Dr. Ibrahim) indicate that antibody-based therapy is a promising strategy to treat mucormycosis. This technology is based on the important discovery that the fungal cell surface proteins encoded by CotH facilitate disease progression by allowing R. oryzae to invade mammalian cells via binding to Glucose Regulated Protein 78 (GRP78), a heat shock conserved protein expressed on endothelial cells lining blood vessels during mucormycosis. Importantly, CotH proteins were found to be conserved among Mucorales (organisms that cause mucormycosis) and absent from any other cell type including mammalian. Our data also show that CotH proteins are key determinants of mucormycosis pathogenesis since R. oryzae coth null mutants have markedly reduced virulence in mouse models of mucormycosis. Further, polyclonal antibodies targeting CotH are highly protective against murine mucormycosis caused by R. oryzae, Mucor, Lichtheimia, Cunninghamella, Rhizomucor, and Apophysomyces. Importantly and highly relevant to this application, anti-CotH murine monoclonal antibodies (mAb) raised against a peptide predicted to be present in the binding domain to GRP78, prevent the ability of R. oryzae to invade and injure endothelial cells in vitro and protect mice from mucormycosis caused by several Mucorales to levels that exceed those seen with antifungal therapy. While these mAbs are a promising new therapy for mucormycosis, the feasibility of further clinical development will hinge upon successful humanization of the Abs. Mouse mAbs cannot be used to treat humans, because humans mount an immune reaction to mouse mAbs that can cause rapid removal of the mAbs, systemic inflammation, severe allergic reactions, and even a risk for death. The humanization process prevents these undesirable effects. Thus, we propose two AIMS: 1) Develop our lead murine mAb into a humanized version with retained/enhanced binding ability to CotH proteins; and 2) Determine the protective activity of the humanized Ab in vitro/in vivo and evaluate its toxicity to human tissues. We propose conservative feasibility milestones that are part of a standard, methodical development pathway for our unique mAbs as a novel treatment for mucormycosis. The proposed work will identify a lead humanized Ab that will go into further development to ultimately test in clinical trials as an adjunctive therapy.

Public Health Relevance Statement:
Mucormycosis is a life-threatening fungal infection that afflicts patients with weakened immune system. Current treatment fail in >50% of patients despite current intervention with antibiotics and surgical removal of destroyed tissues. We will develop a novel antibody therapy that is proven in mice to enhance the efficiency of the current therapy.

NIH Spending Category:
Biotechnology; Clinical Research; Emerging Infectious Diseases; Immunization; Immunotherapy; Infectious Diseases; Orphan Drug; Prevention; Rare Diseases

Project Terms:
Adrenal Cortex Hormones; Affinity; Allergic Reaction; Amino Acids; Angioinvasion; Animal Model; Antibiotics; Antibodies; Antibody Therapy; Antifungal Agents; Antifungal Therapy; Antigens; Attenuated; base; Binding; Biological Sciences; Blood Vessels; Brain; cancer transplantation; cell injury; Cell Line; Cell Surface Proteins; cell type; cGMP production; Clinical; clinical development; Clinical Trials; clinically relevant; combat; cross reactivity; Cunninghamella; Data; Development; Diabetes Mellitus; Diabetic Ketoacidosis; Diagnostic; Disease; Disease Progression; Dose; driving force; effective therapy; Endothelial Cells; Enzyme-Linked Immunosorbent Assay; Erythrocytes; Excision; Fc Immunoglobulins; Feasibility Studies; Funding; Genes; glucose-regulated proteins; Goals; Grant; Heat shock proteins; Heat-Shock Response; Hematogenous; Hematopoietic Stem Cell Transplantation; Human; Human Cloning; human tissue; humanized monoclonal antibodies; IgG1; Immune system; Immunocompromised Host; immunoreaction; Immunotherapy; improved; In Vitro; in vitro activity; in vivo; Incidence; Infection; Inflammation; injured; Injury; Intervention; Invaded; Investments; Iron; Laboratories; Lead; lead candidate; Life; Mammalian Cell; Mediating; method development; Molds; Monoclonal Antibodies; mortality; mortality risk; mouse model; Mucor; Mucorales; Mucormycosis; murine monoclonal antibody; Mus; mutant; Mycoses; Necrosis; Neutropenia; novel; novel therapeutics; operation; Operative Surgical Procedures; Organ Transplantation; Organism; Orphan Drugs; Outcome; Pathogenesis; Pathway interactions; Patients; Penetration; Peptides; Phagocytosis; Pharmaceutical Preparations; pharmacokinetics and pharmacodynamics; Phase; polyclonal antibody; posaconazole; Positioning Attribute; pre-clinical; Prevalence; prevent; Prevention strategy; primary endpoint; Privatization; Process; programs; Proteins; Reporting; Research; Resources; Rhizomucor; Rhizopus; Risk; Risk Factors; secondary endpoint; Serum; Small Business Innovation Research Grant; Small Business Technology Transfer Research; Soldier; Stains; synergism; Technology; Testing; Therapeutics for Rare and Neglected Diseases; Thrombosis; Tissues; Toxic effect; Trauma; treatment strategy; Umbilical vein; United States National Institutes of Health; Vaccines; Virulence; Work; Zygomycosis