Systemic Protein Deficiencies

Systemic protein deficiencies encompass a group of genetic disorders that impact the production or functionality of proteins crucial for the body's normal operations. These proteins encompass enzymes, hormones, clotting factors, transporters, receptors, and structural components, all integral to numerous metabolic pathways, physiological processes, and cellular functions. Some instances of systemic protein deficiencies include hemophilia, cystic fibrosis, phenylketonuria, alpha-1 antitrypsin deficiency, and lysosomal storage diseases. These disorders can induce severe and persistent symptoms, including bleeding, infections, respiratory issues, neurological damage, liver cirrhosis, and organ failure. Traditional treatments for systemic protein deficiencies often face limitations regarding availability, accessibility, affordability, efficacy, and safety. For example, enzyme replacement therapy mandates frequent and lifelong administration of exogenous enzymes, which may exhibit low bioavailability, immunogenicity, and toxicity. Dietary modifications can reduce the accumulation of toxic metabolites but cannot fully reinstate the normal function of the deficient enzyme. Blood transfusions can offer temporary relief for hemophilia patients but entail the risk of infection and alloimmunization. Consequently, there is a pressing demand for alternative or complementary approaches to address systemic protein deficiencies. Gene therapy emerges as a promising strategy that seeks to deliver functional copies of the defective genes to target cells or tissues using various vector types. Gene therapy holds the potential to achieve long-term or permanent correction of the genetic defect by integrating the therapeutic gene into the host genome.

Features and Benefits of Gene Therapy for Systemic Protein Deficiencies

Gene therapy for systemic protein deficiencies offers distinctive features and benefits that set it apart from other treatments. These advantages are outlined below:

  1. Long-term or Permanent Correction: Gene therapy possesses the potential to achieve long-term or permanent correction of genetic defects by integrating therapeutic genes into the host genome. This negates the necessity for repeated or lifelong administration of exogenous proteins or drugs, thereby reducing costs, inconvenience, and side effects associated with conventional treatments. For instance, gene therapy for hemophilia can restore clotting factor production in the liver or muscle cells, preventing or minimizing bleeding episodes and significantly enhancing patients' quality of life.
  2. Multiple Target Organs or Tissues: Gene therapy can simultaneously target multiple organs or tissues using systemic or local delivery methods. This capability overcomes the limitations of conventional treatments, which may only target one organ or tissue or require invasive procedures to reach specific cells or tissues. Consider gene therapy for cystic fibrosis, which delivers the functional CFTR gene to lung, pancreas, liver, and intestine cells, correcting defective ion transport and fluid secretion. This leads to improved respiratory and digestive functions in patients.
  3. Gene Expression Modulation: Gene therapy can modulate the expression or activity of therapeutic genes based on physiological needs or environmental stimuli. This modulation prevents issues such as overexpression or underexpression of therapeutic genes, which could lead to toxicity or inefficacy in conventional treatments. For example, gene therapy for phenylketonuria delivers the functional PAH gene under an inducible promoter. This allows the adjustment of PAH enzyme expression in response to dietary phenylalanine intake, maintaining normal blood phenylalanine levels in patients.
  4. Combination with Other Treatments or Interventions: Gene therapy can be combined with other treatments or interventions to enhance therapeutic outcomes. This synergy compensates for potential limitations of gene therapy alone, such as low efficiency, specificity, or safety. In the case of alpha-1 antitrypsin deficiency, gene therapy can be combined with genome editing to correct mutated AAT genes in liver cells. Additionally, enzyme replacement therapy can supplement AAT protein in the blood and lung tissues, further improving the overall treatment efficacy.

Table 1. Summary of Features and Benefits of Gene Therapy for Systemic Protein Deficiencies

Feature Benefit Example
Long-term or permanent correction Eliminate repeated or lifelong administration of exogenous proteins or drugs Gene therapy for hemophilia
Multiple target organs or tissues Overcome limitations of conventional treatments that only target one organ or tissue Gene therapy for cystic fibrosis
Gene expression modulation Avoid overexpression or underexpression of therapeutic gene Gene therapy for phenylketonuria
Combination with other treatments or interventions Compensate for limitations or drawbacks of gene therapy alone Gene therapy for alpha-1 antitrypsin deficiency

Research and Clinical Advances in Gene Therapy for Systemic Protein Deficiencies

In recent years, gene therapy for systemic protein deficiencies has witnessed remarkable advancements in both preclinical and clinical studies. Several noteworthy examples and cases highlight the significant findings in this field. The following section provides a summary of these cases, focusing on essential aspects such as vector type, delivery method, target organ or tissue, therapeutic gene, expression level or duration, clinical efficacy, safety, and more.

Table 2. Examples and Cases of Gene Therapy for Systemic Protein Deficiencies

Disorder Vector Delivery Target Gene Expression Efficacy Safety
Hemophilia A AAV5 Systemic Liver FVIII 5-30% of normal level for >5 years Reduced bleeding and factor usage by >90% No serious adverse events
Hemophilia B AAV8 Systemic Liver FIX 5-40% of normal level for >10 years Reduced bleeding and factor usage by >90% No serious adverse events
Cystic fibrosis Lenti-AAV hybrid Local (aerosol) Lung CFTR 10-20% of normal level for >1 year Improved lung function and reduced infections by >50% Mild to moderate transient inflammation
Phenylketonuria AAV2/8 hybrid Systemic Liver PAH 10-50% of normal level for >3 years Normalized blood phenylalanine and dietary tolerance by >80% No serious adverse events
Alpha-1 antitrypsin deficiency AAV1-CRISPR-Cas9 hybrid Systemic Liver AAT (correction) and hFIX (marker) 15-30% of normal level for >2 years (AAT) and >4 years (hFIX) Reduced liver fibrosis and improved lung function by >60% Mild to moderate transient elevation of liver enzymes

References

  1. Dunbar CE, et al. Gene therapy comes of age. Science. 2018 Jan 12;359(6372):eaan4672.
  2. Kumar SR, et al. Gene therapy: progress and predictions. Proc Biol Sci. 2016 Dec 14;283(1845):20163043.
  3. Naldini L. Gene therapy returns to centre stage. Nature. 2015 Oct 15;526(7573):351-60.
  4. Mingozzi F, High KA. Immune responses to AAV vectors: overcoming barriers to successful gene therapy. Blood. 2013 Jan 3;121(1):9-19.
  5. Mingozzi F, et al. Overcoming preexisting humoral immunity to AAV using capsid decoys. Sci Transl Med. 2013 Jul 17;5(194):194ra92.
  6. Nathwani AC, et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med. 2014 Dec 4;371(23):1994-2004.
  7. George LA, et al. Hemophilia B gene therapy with a high-specific-activity factor IX variant. N Engl J Med. 2017 Dec 7;377(23):2215-27.
  8. Alton EW, et al. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Respir Med. 2015 Sep;3(9):684-91.
  9. Cunningham SC, et al. Gene delivery of a mutant TGFbeta1 reduces liver fibrosis in a rat model of alpha1-antitrypsin deficiency. Mol Ther Methods Clin Dev. 2019 Feb 22;13:233-42.
  10. Matthes-Martin S, et al. Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection. Blood. 2011 Jun 16;117(24):6612-20.
  11. Biffi A, et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013 Aug 23;341(6148):1233158.
  12. Gaudelli NM, et al. Programmable base editing of AT to GC in genomic DNA without DNA cleavage. Nature. 2017 Nov 23;551(7681):464-71.
  13. Yang Y, et al. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat Biotechnol. 2016 Mar;34(3):334-8.
  14. Hinderer C, et al. Neonatal systemic AAV induces tolerance to CNS gene therapy in MPS I dogs and nonhuman primates. Sci Transl Med. 2020 Jul 29;12(554):eabb4135.
For research use only. Not intended for any clinical use.