Tuesday, August 4, 2015

Evolution of Treatments- Then and Now (2014)

Hemophilia is a bleeding disorder.  In a hemophiliac blood clots significantly slower than a normal person.  Due to this blood accumulates in the local tissue internally until the clot forms.  This is noticeable primarily in joints but can occur in muscle, soft-tissue or within an organ (GI, intracranial).  If a bleed is not stemmed, it can be fatal. Hemophilia is an incurable condition today- historically considered transmitted via inheritance of a faulty gene by the male child from the mother.  However, over the last decade or so, hemophilia has been observed in families with no prior incidence.  This has been attributed to spontaneous gene mutation.  Moreover, there are sporadic cases of female hemophiliacs in the general population.
A number of proteins called 'Factors' work in tandem to form a blood clot.  The sequence of their actions is defined in what is known as the Coagulation Pathway or Cascade.  The Factors are designated by Roman Numerals ( I, II... , VIII, IX... and such).  A missing, inadequate or incomplete Factor due to the faulty gene can cause an unstable clot and a bleeding disorder.  Hemophilia is caused by a defect in the Factor VIII (FVIII,in Hemophilia A)  or Factor IX (FIX, Hemophilia B) gene leading to various levels of the circulating clotting protein.  Hemophilia is classified based on the levels of Factor as severe( < 1% of normal), moderate (1-5%) or mild (5-40%).
Historically, hemophilia has been treated by replacing the missing Factor, administered intravenously via an injection.  Treatments have evolved over the years starting with matched fresh blood, purified pooled plasma, fractionated plasma, freeze-dried concentrates (cryoprecipitate), and monoclonal antibody (mAb) purified protein.  The simple difference between these products was noticeable in the volume used and their purity.  In the 80s a large number of patients were infected with HIV and Hepatitis B and Hepatitis C from the use of pooled plasma products.  Safety of Factor was suspect. The next milestone, a significant step forward, in the treatment for hemophilia came from to the evolution of gene cloning and recombinant technology.  in 1992, FDA approved the first recombinant Factor VIII product for clinical use.  This antihemophilic Factor is a glycoprotein synthesized by a genetically engineered Chinese Hamster Ovary (CHO) cell line, followed by several stages of immunoaffinity chromatography including mAb directed to FVIII.  All manufacturers of FVIII jumped on the recombinant bandwagon and developed their own products.  The differences were primarily either in the active Factor domain expression or with the steps in the purification process.  Until the early 2000s these were the purest form of injectable Factor for clinical use.
The landmark availability of recombinant FVIII had one caveat.  All available Factors were using some form of human albumin in their manufacture.  This was present in the cell culture medium or used as a protein stabilizer.  The human albumin offered a theoretical chance of viral transmission and hence a safety concern.  The next generation of FVIII, approved in 2003, was the first human plasma/albumin free product.  This has been deemed the safest product in the hemophilia market.
While the safety concerns were being addressed by the laboratories and the pharmaceuticals, the overall patient care systems were also evolving.  Care that once required hospitalization or an out-patient visit had become home-based therapy.  A number of home-care pharmacy businesses became the middle-tier between manufacturers and patients. Patients were getting treatment from a local general physician or hematologist to a federally funded hemophilia treatment center (HTC).  The treatment regimen also changed from on-demand (episode based) to prophylactic (preventive) schedule of infusions.  Care went from specialist- patient interaction to a multi-disciplinary comprehensive care for hemophilia.  Hemophilia camps taught children to self-infuse at an early age.
With the safety concerns of antihemophilic products behind them the trend in the past decade has been to prolong the availability of active protein in circulation.  This is technically measured in terms of half-life (T 1/2) of the protein.  Half-life is the time required for the protein to reach half its original activity level.  For FVIII the T 1/2 is 8-12h.  To be effective the circulating Factor should be maintained at or above a certain level.  This is commonly termed the 'trough' value.  The trough various for different individuals based on their daily activity.  A 10% level is considered a good trough.  In order to maintain this Factor level a patient on prophylaxis has to infuse three times a week at the basic dose of 20U/kg body-weight.
This next advance in Factor therapy addressed the longevity of Factor in vivo.  Recently, in 2014, FVIII and FIX products with extended half-life was available.  These products had 1.5 -3.0 times longer half-life compared to current products.  Fc fusion-based technology, previously used with other drugs, was used in these Factor products.  This approach combined two molecular structures- a single molecule of recombinant Factor fused with the dimeric Fc domain of IgG1- was used to extend the half-life in a clinically usable way.
Treatment for hemophilia has come a long way over the last decades.  Progress has been made in terms of improvements in the volume, safety, storage temperature, portability and dosing regimen.  Simultaneously, various infusion devices have also been developed addressing the 'accidental pricks' danger to the patient or healthcare provider.  Several types of reconstitution devices are in the market, typically packaged with the Factor product.  The product development continues with more manufacturers entering the market with a dozen or more products in the pipeline or various phase of clinical trials.  Recent successes have been reported with gene therapy trials for Factor IX.


See also
Hemophilia Therapies in the Pipeline (2015-)

Hemophilia Therapies In the pipeline (2015-)


Hemophilia is a bleeding disorder caused by defective proteins, also called Factor, in the blood clotting cascade.  Treatment for hemophilia has traditionally been through intravenous injections of adequate concentrations of the missing protein.  At this time there is no cure for hemophilia, however, the condition is well controlled by such treatment.  Recently, the replacement therapies have focussed on extending the half-life of Factors.  Today there is one such Factor VIII (FVIII) and Factor IX (FIX) product available in the market.  Eloctate for FVIII and Alprolix for FIX were approved in 2014 as the first extended half-life (EHF) product to market from Biogen-Idec.  Baxter introduced Obizur (FVIII) to treat acquired hemophilia in 2015.   This is a recombinant porcine FVIII.  Baxter is awaiting FDA approval on a new EHF FVIII product- Adynovate (BAX855) a pegylated molecule.  This is a full-length rFVIII protein.

There are a number of longer lasting Factor products in late phases of clinical trials or awaiting FDA approval.  Biotechnology companies along with pharmaceuticals are actively developing and testing new molecules for their potential use to treat hemophilia.  These novel approaches to Factor development fill the pharmaceutical pipeline.  Additional molecules with extended half-life using pegylation or fusion to albumin are in clinical trials. 

Various mechanisms to promote the formation of a stable clot are being investigated. A humanized bispecific monoclonal antibody (mAb) to FIXa and FX (ACE910) from Genentech is entering Phase 2 study. This is a small molecule and can be administered subcutaenously.  Such a molecule provides two very significant advantage. First, it is a benefit to patients with venous access issues; the other it bypasses the need for FVIII by stiumulating the rate of FXa generation via the assembly of FIX1 and FX. The latter is a clear benefit to treat patients with inhibitors to FVIII. N9-GP is a EHF FIX product (NOVO Nordisk) is in Phase 3 clinical trials.  N9-GP, a glycopegylated molecule, shows pharmacokinetics data (pK) with a half-life of 110 hours.  CSL-Behring has submitted for FDA approval on a rFIX albuimin fusion protein  (rFIX-FP) for use in hemophilia B. a rFVIIa-Albumin fusion protein (CSL Behring), a rVWF-Albumin fusion protein and a rFVIIa-CTP fusion protein (Prolor Biotech, Israel).

Four proteins- TFPI, protein C, protein S, ATIII - act as brakes in the coagulation cascade.  Researchers are trying to takes these brakes off as a mechanism to bypass the FVIII, FIX requirement.  These are currently being studied in animals.  Neutralizing monolclonal antibody (mAb) negates the effect of TFPI  on the extrinisic pathway activation. A Novo Nordisk product concizumab (mAb2021) demonstrates hemostatic effect by blocking the TFPI action on the active site of FXa in monkeys after subcutaneous administration.  Reduction in thrombin formation has been attempted by suppressing the hepatocytes production of antithrombin (AT) mRNA via subcutaneous injection. Alnylam Pharmaecuticals reported a phase I study of a ALN-AT3 (short interfering RNA product), targeting thrombin.  

Hemophilia has always been an attractive candidate for gene therapy, being a single gene abnormality.   Gene therapy can also provide a lasting effect with a single treatment and become a cost-effective alternative to current treatments.  Adeno-associated virus (AAV) or lentiviral vectors have been used for hemophilia.  AAV is a single-stranded parvovirus and has the advantage of low immunogenicity, high safety and transferability to quiescent cells.  AAV Serotype 8 (AAV8) allows gene expression in the liver, where coagulation factors are produced in animal studies. Nathwani et al recently reported therapeutic FIX expression for 3 years with AAV8 gene therapy in humans.  Clinicaltrials.gov gave 7 hits on a search with "AAV + Hemophilia" in the criteria.   Lentiviruses are low-risk vector compared to AAV.  Lentiviruses are more efficient at transducing quiescent cells such as the hematopoietic stem cells than retroviral vectors.  Lentiviral vectors have been used to transfer genes into hepatocytes by direct injection of the vector.  The successes of gene therapy for FIX is not easily translated for FVIII due to the larger size of the FVIII gene (7 kbps).  This will require an efficient high-yielding delivery system to achieve therapeutic levels.  Glybera®is a commercial AAV vector approved in Europe.

Cell therapy involves transplanting and induction of Factor expressing cells.  Cells such as mesenchymal stem cells, endothelial cells, hepatocytes are transduced with a Factor carrying vector such as a plasmid or a virus.  The advantage is that these cells do not get disseminated in the body and the risk of triggering a anti-vector immune response is reduced.  The majority of the studies on gene therapy and cell therapy are currently done in animal.  This is because of an ideal non-human primate model of the disease state.  Very recently hemophilia A pigs are being studied as a posible model.

Genome engineering is another area where rapid advancement is taking place.  Gene-editing tools such as the CRISP/Cas9 system, Transcription activator-like effector neucleses (TALENs) and zinc finger nucleases, could have future application in hemophilia gene therapy.

Therapy for hemophilia has come a long way after the discovery of cryprecipitate to treatment.  The landscape of the standard replacement therapy is likely to be supplemented by the new EHF Factor.  A number of clinical trials are being conducted with alternate routes, Factor bypassing agents, disrupting anti-thrombin activity, gene and cell therapy. 



Additional acronyms:
CTP = C-terminal peptide; F = factor; FcIg = Fc domain of immunoglobulin (Ig)G; kDa = kilodaltons; mAB = monoclonal antibody; mRNA = messenger RNA; PEG = polyethylene glycol; siRNA = small interfering RNA; T1/2 = circulating half-life; TFPI = tissue factor pathway inhibitor; vWF = von Willebrand factor

See also