treat chronic diseases

INFEPERIUM™

A breakthrough Immunotherapy and why it is different!

WARNING!!! This page is for licensed physicians and medical researchers. See disclaimer below.

The Science Behind INFEPERIUM™

In simple terms INFEPERIUM™ is a Biological Response Modifier (BRM) / Immunomodulator that helps reset the immune system so your body can heal itself.  It addresses in an endogenous (from inside the body) way the majority causes of Chronic Disease and their effects on the body.

The active ingredient in INFEPERIUM™ modulates both the adaptive and innate immune systems (without negative side effects) and allows the body to eliminate chronic inflammation, a major cause in all diseases. This is the same active ingredient that is in our USDA approved immunomodulator.

INFEPERIUM™ restores proper cell signaling, pathogen recognition and proper cell energy, Adenosine Triphosphate production (ATP) endogenously.

INFEPERIUM™ is a major breakthrough in Immunotherapy. The action of INFEPERIUM™ is to modulate both the innate and adaptive immune systems that have been degraded and compromised due to disease, environmental, bacterial, viral etc. to normalization and homeostasis allowing the bodies own healing and defense mechanisms to restore proper cell signaling, proper cellular respiration versus fermentation, regulation of cellular transport, and healing. INFEPERIUM™ embodies the latest and most medically accepted theories and practices of immunotherapy, which is now at the forefront and becoming the standard of care for chronic diseases which include cancer, a wide range of autoimmune conditions, and most importantly addressing the growing concern of bacterial resistance.

What INFEPERIUM™’s breakthrough Immunotherapy can do, and why it is different.

INFEPERIUM™’s immunomodulator is one of the few molecules, if not the only one known today, that affects and modulates both the innate and adaptive immune systems without negative side effects.

The primary triggers for Chronic Disease are:

The establishment of a state of Chronic Inflammation (CI), which is now considered the main cause of the vast majority of disease.

Due to the prolonged state of CI, improper and aberrant cellular signaling begins which is the harbinger of most AutoImmune(AI) conditions.

This state of cellular "Misinformation" creates over and under reaction of the immune response and inhibits the bodies defense mechanisms from properly functioning. This includes but is not limited to depressed CD4 and CD8 counts. 

Suppressed MHC I and MHC II, which Marshall the forces of T cells, B cells, NK cells and various Macrophages, that prevent pathogens from invading the bodies systems or once they have attacked them to consume and destroy them. Most importantly interfering with the Jak-Stat signaling pathway which controls immune deficiency syndromes and most Cancers.
             
These three states determine the vast majority of disease. However, if you can address these above states of disease from an Endogenous state, you are going to able to reverse a vast number of diverse conditions that are created by these altered states of health. As well as bringing the patient to Homeostasis.

INFEPERIUM™ addresses these altered states by the modulation of the bodies cytokine cascade from attacking healthy cells to reversing GI and inducing proper cell signaling and pathogen recognition. That it modulates and reestablishes proper function of the innate and adaptive immune systems and responses is the key to the unique effectiveness of INFEPERIUM™.

The Science

Why Biologics?
According to the United States Food & Drug Administration (FDA) “Biological products include a wide range of products such as vaccines, blood and blood components, allergenics, somatic cells, gene therapy, tissues, and recombinant therapeutic proteins. Biologics can be composed of sugars, proteins, or nucleic acids or complex combinations of these substances, or may be living entities such as cells and tissues. Biologics are isolated from a variety of natural sources - human, animal, or microorganism - and may be produced by biotechnology methods and other cutting- edge technologies. Gene-based and cellular biologics, for example, often are at the forefront of biomedical research, and may be used to treat a variety of medical conditions for which no other treatments are available.”


According to the FDA, how do biological products differ from conventional drugs?
“In contrast to most drugs that are chemically synthesized and their structure is known, most biologics are complex mixtures that are not easily identified or characterized. Biological products, including those manufactured by biotechnology, tend to be heat sensitive and susceptible to microbial contamination. Therefore, it is necessary to use aseptic principles from initial manufacturing steps, which is also in contrast to most conventional drugs.”


Again, according to the FDA, “Biological products often represent the cutting-edge of biomedical research and, in time, may offer the most effective means to treat a variety of medical illnesses and conditions that presently have no other treatments available.” 

Therefore, Biologic response modifiers (BRMs) can improve or modify the body's natural response to infection and disease. BRMs, a drug class of their own, are the newest medications which are based on compounds made by living cells, not synthetic drugs which the body naturally attempts to reject which frequently leads to injury of major organs like the liver and kidneys.

Biological response modifiers (BRMs) are substances that modify immune responses. They can be both endogenous (produced naturally within the body) and exogenous (from outside the body, as pharmaceutical drugs), and they can either enhance an immune response or suppress it.

Some of these substances arouse the body's response to an infection, and others can keep the response from becoming excessive. Thus they serve as immunomodulators in immunotherapy (therapy that makes use of immune responses), which can be helpful in treating many diseases, such as cancer (where targeted therapy often relies on the immune system being used to attack cancer cells) and in treating autoimmune diseases (in which the immune system attacks the self), such as some kinds of arthritis and dermatitis.

Most BRMs are biopharmaceuticals (biologics), including monoclonal antibodies, interleukin 2, interferons, and various types of colony-stimulating factors (e.g., CSF, GM-CSF, G-CSF). "Immunotherapy makes use of BRMs to enhance the activity of the immune system to increase the body's natural defense mechanisms against cancer”, whereas BRMs for rheumatoid arthritis aim to reduce inflammation.

References:
1. He Y, Shimoda M, Ono Y, Villalobos IB, Mitra A, Konia T, Grando SA, Zone JJ, Maverakis E (2015). "Persistence of Autoreactive IgA-Secreting B Cells Despite Multiple Immunosuppressive Medications Including Rituximab". JAMA Dermatol 151 (6): 646–50. doi:10.1001/jamadermatol.2015.59. PMID 25901938.
2. Tzianabos AO (2000). "Polysaccharide immunomodulators as therapeutic agents: structural aspects and biologic function.". Clin Microbiol Rev 13 (4): 523–33. doi:10.1128/CMR.13.4.523-533.2000. PMC 88946. PMID 11023954.
3. "The Riordan IVC Protocol for Adjunctive Cancer Care: Intravenous Ascorbate as a Chemotherapeutic and Biological Response Modifying Agent" (PDF). Riordan Clinic Research Institut. February 2013. Retrieved 2 February 2014.
4. "High-Dose Vitamin C (PDQ®): Human/Clinical Studies". National Cancer Institute. Retrieved 2 February 2014.
5. Deepak A. Rao; Le, Tao; Bhushan, Vikas. First Aid for the USMLE Step 1 2008 (First Aid for the Usmle Step 1). McGraw-Hill Medical. ISBN 0-07-149868-0.

Immunotherapy and Immunomodulators:
Immunotherapy is the treatment of disease by inducing, enhancing or suppressing an Immune Response. Jedd Wolchok MD summed up Immunotherapy just last year in response to Immunotherapy being Science Magazines 2013 Breakthrough of the Year. “10 years ago Immunotherapy was almost in the category of snake oil. A few years ago it was stated that Immunotherapy can work. Now Immunotherapy has entered the main stream.”

James T. Allison, Phd and professor and Chair of Immunology at the MD Anderson Cancer Research Institute (CRI) has received the prestigious Tang Prize, the Louisa Gross Horwitz Prize and the Harvey Prize, all in 2014 for his work on CTLA-4 as an antibody inhibitor in many treatment resistant cancers with amazing survival statistics. Work in CTLA-4 & PD-1 also called “immune check point therapies” are the only two presently approved FDA ImmunoTherapy agents so far.

Why Immunotherapy?
Specificity. T-cells recognize peptides, which are short chain biological molecules that are produced by every cell category including virus, bacteria, cancer/mutations. T-cells can recognize them and can destroy them, and most importantly they have the following two important elements.
First, Memory. As T-cells destroy pathogens, which are biological or environmental invaders which produce abnormal cells, they go through 3 phases: expansion, contraction and memory.

  • Expansion is when the T-cells multiply to overwhelm the attacker
  • Contraction is when they diminish due to program cell death after the attacker is neutralized.
  • Memory is a multiplication of cells that remember the genetic code of the attacker. Memory cells proliferate and circulate through the body standing ready to attack.


Second, Adaptability. Cancerous tumors, viruses and bacteria, can mutate and alter themselves in order to evade various medical therapies. Cancer alone has 9 resistant mechanisms used to trick, hide and survive. So called present “Standard of Care Therapies” which use in large measure synthetically produced drugs frequently harmful to the body, but can’t adapt fast enough to counteract and destroy the mutation capabilities of cancer cells, bacteria and viruses. However, T-cell antigen receptors can. They adapt and respond as quickly as the pathogens, because they can combine and alter themselves as many 10 to the 15th power in different combinations. The stimulation of the T-cells is a natural response of the body’s immune system to the injection of INFEPERIUM™. This is the key to why and how of immunotherapy.

Immunomodulators:
An immunomodulator is a substance that either suppresses or activates the body's immune response. These substances are separated into two groups: immunosuppressants and immune activators. Immunosuppressants inhibit the body’s natural immune response, while immune activators generally condition or reprogram it to target a specific disease-causing agent.


Immunomodulators can be produced in synthetic form or naturally in the body. Cytokines are examples of innate immune mediators. Synthetic versions are available in either immunosuppressant or immune activator forms. A suppressant immunomodulator works by inhibiting the activation of critical immune system agents such as calcineurin and the formation of thymus cells (T-cells) and antibodies. In comparison, an activating immunomodulator uses the process of adaptive immunity to recondition lymphocytes and T-cells to kill known pathogens or tumor cells.


Cyclosporine and methotrexate are commonly used synthetic immunosuppressors. Methotrexate is used in patients with autoimmune ailments. Lupus and rheumatoid arthritis are examples of autoimmune disorders that cause the patient's body to attack his or her own cells. Eventually the targeted cells and tissue become damaged after repeated attacks.

The process of organ rejection is similar to autoimmune dysfunction, except the immune system targets the transplanted organ rather than the body’s own cells. Organ transplant recipients take suppressant drugs such as cyclosporine, tacrolimus and sirolimus to prevent organ rejection. Nearly all transplant recipients, except a rare few, must adhere to a strict daily regime that involves taking these medications for life. Not taking the medications as prescribed will almost always induce organ rejection, which could lead to death. Due to the medication’s toxic side effects, immuno-suppressors should only be used in cases of severe autoimmune dysfunction or organ transplantation.

Immunomodulators that activate the immune system include vaccines and cancer immunotherapy. Vaccines work by exposing the patient to weakened or inactive forms of certain bacteria and viruses. The immune system then adapts by producing antibodies that are programmed to immediately kill the introduced pathogen once it re-enters the body, which is called adaptive immunity.


Cancer immunotherapy is very similar to pathogen vaccination. The difference between the two therapies is the agent in which adaptive immunity is induced. Vaccines use microorganisms, while cancer immunotherapy uses microorganisms and enhanced immune cells. Microorganism-based cancer immunotherapies are used to combat some forms of cervical and liver cancers caused by viruses. A cell-based immunomodulator, on the other hand, uses enhanced immune cells such as cytotoxic T lymphocytes (CTLs), dendritic cells (DC) and natural killer cells (NK cells) to target and destroy the patient’s cancerous cells.

References:
1. "immunotherapies definition". Dictionary.com. Retrieved 2009-06-02.
2.Masihi KN (July 2001). "Fighting infection using immunomodulatory agents". Expert Opin Biol Ther 1 (4): 641–53. doi:10.1517/14712598.1.4.641. PMID 11727500.
3.Rosenberg SA (January 1984). "Adoptive immunotherapy of cancer: accomplishments and prospects". Cancer Treat Rep 68 (1): 233–55. PMID 6362866.
4.Yang Q, Hokland ME, Bryant JL, Zhang Y, Nannmark U, Watkins SC, Goldfarb RH, Herberman RB, Basse PH (July 2003). "Tumor-localization by adoptively transferred, interleukin-2-activated NK cells leads to destruction of well-established lung metastases". Int. J. Cancer 105 (4): 512–9. doi:10.1002/ijc.11119. PMID 12712443.
5. Egawa K (2004). "Immuno-cell therapy of cancer in Japan". Anticancer Res. 24 (5C): 3321–6. PMID 15515427.
6. Li K, Li CK, Chuen CK, Tsang KS, Fok TF, James AE, Lee SM, Shing MM, Chik KW, Yuen PM (February 2005). "Preclinical ex vivo expansion of G-CSF-mobilized peripheral blood stem cells: effects of serum-free media, cytokine combinations and chemotherapy". Eur. J. Haematol. 74 (2): 128–35. doi:10.1111/j.1600-0609.2004.00343.x. PMID 15654904.
7. Fujita K, Ikarashi H, Takakuwa K, Kodama S, Tokunaga A, Takahashi T, Tanaka K (May 1995). "Prolonged disease-free period in patients with advanced epithelial ovarian cancer after adoptive transfer of tumor-infiltrating lymphocytes". Clin. Cancer Res. 1 (5): 501–7. PMID 9816009.
8. Kimura H, Yamaguchi Y (July 1997). "A phase III randomized study of interleukin-2 lymphokine-activated killer cell immunotherapy combined with chemotherapy or radiotherapy after curative or noncurative resection of primary lung carcinoma". Cancer 80 (1): 42–9. doi:10.1002/(SICI)1097-0142(19970701)80:1<42::AID-CNCR6>3.0.CO;2-H. PMID 9210707.
9. Takayama T, Sekine T, Makuuchi M, Yamasaki S, Kosuge T, Yamamoto J, Shimada K, Sakamoto M, Hirohashi S, Ohashi Y, Kakizoe T (September 2000). "Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial". Lancet 356 (9232): 802–7. doi:10.1016/S0140-6736(00)02654-4. PMID 11022927.
10. Kono K, Takahashi A, Ichihara F, Amemiya H, Iizuka H, Fujii H, Sekikawa T, Matsumoto Y (June 2002). "Prognostic significance of adoptive immunotherapy with tumor-associated lymphocytes in patients with advanced gastric cancer: a randomized trial". Clin. Cancer Res. 8 (6): 1767–71. PMID 12060615.
11. a b Järvinen R, Kaasinen E, Sankila A, Rintala E (August 2009). "Long-term efficacy of maintenance bacillus Calmette-Guérin versus maintenance mitomycin C instillation therapy in frequently recurrent TaT1 tumours without carcinoma in situ: a subgroup analysis of the prospective, randomised FinnBladder I study with a 20-year follow-up". Eur. Urol. 56 (2): 260–5. doi:10.1016/j.eururo.2009.04.009. PMID 19395154.
12.van Seters M, van Beurden M, ten Kate FJ, Beckmann I, Ewing PC, Eijkemans MJ, Kagie MJ, Meijer CJ, Aaronson NK, Kleinjan A, Heijmans-Antonissen C, Zijlstra FJ, Burger MP, Helmerhorst TJ (April 2008). "Treatment of vulvar intraepithelial neoplasia with topical imiquimod". N. Engl. J. Med. 358 (14): 1465–73. doi:10.1056/NEJMoa072685. PMID 18385498.
13. Buck HW, Guth KJ (October 2003). "Treatment of vaginal intraepithelial neoplasia (primarily low grade) with imiquimod 5% cream". J Low Genit Tract Dis 7 (4): 290–3. doi:10.1097/00128360-200310000-00011. PMID 17051086.
14. Davidson HC, Leibowitz MS, Lopez-Albaitero A, Ferris RL (September 2009). "Immunotherapy for head and neck cancer". Oral Oncol. 45 (9): 747–51. doi:10.1016/j.oraloncology.2009.02.009. PMID 19442565.
15. Dani T, Knobler R (2009). "Extracorporeal photoimmunotherapy-photopheresis". Front. Biosci. 14 (14): 4769–77. doi:10.2741/3566. PMID 19273388.
16. Eggermont AM, Schadendorf D (June 2009). "Melanoma and immunotherapy". Hematol. Oncol. Clin. North Am. 23 (3): 547–64, ix–x. doi:10.1016/j.hoc.2009.03.009. PMID 19464602.
17. Chuang CM, Monie A, Wu A, Hung CF (2009). "Combination of apigenin treatment with therapeutic HPV DNA vaccination generates enhanced therapeutic anti tumor effects". J. Biomed. Sci. 16 (1): 49. doi:10.1186/1423-0127-16-49. PMC 2705346. PMID 19473507.
18. Pawlita M, Gissmann L (April 2009). "[Recurrent respiratory papillomatosis: indication for HPV vaccination?]". Dtsch. Med. Wochenschr. (in German). 134 Suppl 2: S100–2. doi:10.1055/s-0029-1220219. PMID 19353471.
19. Kang N, Zhou J, Zhang T, Wang L, Lu F, Cui Y, Cui L, He W (August 2009). "Adoptive immunotherapy of lung cancer with immobilized anti-TCRgammadelta antibody-expanded human gammadelta T-cells in peripheral blood". Cancer Biol. Ther. 8 (16): 1540–9. doi:10.4161/cbt.8.16.8950. PMID 19471115.
20. Overes IM, Fredrix H, Kester MG, Falkenburg JH, van der Voort R, de Witte TM, Dolstra H (2009). "Efficient activation of LRH-1-specific CD8+ T-cell responses from transplanted leukemia patients by stimulation with P2X5 mRNA-electroporated dendritic cells". J. Immunother. 32 (6): 539–51. doi:10.1097/CJI.0b013e3181987c22. PMID 19483655.
21. Di Lorenzo G, Buonerba C, Kantoff PW (September 2011). "Immunotherapy for the treatment of prostate cancer". Nature Reviews Clinical Oncology 8 (9): 551–61. doi:10.1038/nrclinonc.2011.72. PMID 21606971.
22. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME (April 2008). "Adoptive cell transfer: a clinical path to effective cancer immunotherapy". Nature Reviews Cancer 8 (4): 299–308. doi:10.1038/nrc2355. PMC 2553205. PMID 18354418.
23. Motohashi S, Nakayama T (2009). "Natural killer T cell-mediated immunotherapy for malignant diseases". Front Biosci (Schol Ed) 1: 108–16. doi:10.2741/S10. PMID 19482686.
24. Khattar M, Chen W, Stepkowski SM (2009). "Expanding and converting regulatory T cells: a horizon for immunotherapy". Arch. Immunol. Ther. Exp. (Warsz.) 57 (3): 199–204. doi:10.1007/s00005-009-0021-1. PMID 19479206.
25. Rosenberg SA, Aebersold P, Cornetta K, Kasid A, Morgan RA, Moen R, Karson EM, Lotze MT, Yang JC, Topalian SL (August 1990). "Gene transfer into humans--immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction". N. Engl. J. Med. 323 (9): 570–8. doi:10.1056/NEJM199008303230904. PMID 2381442.
26. Antony PA, Piccirillo CA, Akpinarli A, Finkelstein SE, Speiss PJ, Surman DR, Palmer DC, Chan CC, Klebanoff CA, Overwijk WW, Rosenberg SA, Restifo NP (March 2005). "CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells". Journal of Immunology 174 (5): 2591–601. doi:10.4049/jimmunol.174.5.2591. PMC 1403291. PMID 15728465.
27. Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, Hwang LN, Yu Z, Wrzesinski C, Heimann DM, Surh CD, Rosenberg SA, Restifo NP (October 2005). "Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells". J. Exp. Med. 202 (7): 907–12. doi:10.1084/jem.20050732. PMC 1397916. PMID 16203864.
28. Dummer W, Niethammer AG, Baccala R, Lawson BR, Wagner N, Reisfeld RA, Theofilopoulos AN (July 2002). "T cell homeostatic proliferation elicits effective antitumor autoimmunity". J. Clin. Invest. 110 (2): 185–92. doi:10.1172/JCI15175. PMC 151053. PMID 12122110.
29. Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, Robbins PF, Huang J, Citrin DE, Leitman SF, Wunderlich J, Restifo NP, Thomasian A, Downey SG, Smith FO, Klapper J, Morton K, Laurencot C, White DE, Rosenberg SA (November 2008). "Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens". J. Clin. Oncol. 26 (32): 5233–9. doi:10.1200/JCO.2008.16.5449. PMC 2652090. PMID 18809613.
30. Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, Topalian SL, Sherry R, Restifo NP, Hubicki AM, Robinson MR, Raffeld M, Duray P, Seipp CA, Rogers-Freezer L, Morton KE, Mavroukakis SA, White DE, Rosenberg SA (October 2002). "Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes". Science 298 (5594): 850–4. doi:10.1126/science.1076514. PMC 1764179. PMID 12242449.
31. Pilon-Thomas S, Kuhn L, Ellwanger S, Janssen W, Royster E, Marzban S, Kudchadkar R, Zager J, Gibney G, Sondak VK, Weber J, Mulé JJ, Sarnaik AA (October 2012). "Efficacy of adoptive cell transfer of tumor-infiltrating lymphocytes after lymphopenia induction for metastatic melanoma". J. Immunother. 35 (8): 615–20. doi:10.1097/CJI.0b013e31826e8f5f. PMID 22996367.
32. Manjunath SR, Ramanan G, Dedeepiya VD, Terunuma H, Deng X, Baskar S, Senthilkumar R, Thamaraikannan P, Srinivasan T, Preethy S, Abraham SJ (January 2012). "Autologous immune enhancement therapy in recurrent ovarian cancer with metastases: a case report". Case Rep Oncol 5 (1): 114–8. doi:10.1159/000337319. PMC 3364094. PMID 22666198.
33. Li Y, Zhang T, Ho C, Orange JS, Douglas SD, Ho WZ (December 2004). "Natural killer cells inhibit hepatitis C virus expression". J. Leukoc. Biol. 76 (6): 1171–9. doi:10.1189/jlb.0604372. PMID 15339939.
34. Doskali M, Tanaka Y, Ohira M, Ishiyama K, Tashiro H, Chayama K, Ohdan H (March 2011). "Possibility of adoptive immunotherapy with peripheral blood-derived CD3⁻CD56+ and CD3+CD56+ cells for inducing antihepatocellular carcinoma and antihepatitis C virus activity". J. Immunother. 34 (2): 129–38. doi:10.1097/CJI.0b013e3182048c4e. PMID 21304407.
35. Terunuma H, Deng X, Dewan Z, Fujimoto S, Yamamoto N (2008). "Potential role of NK cells in the induction of immune responses: implications for NK cell-based immunotherapy for cancers and viral infections". Int. Rev. Immunol. 27 (3): 93–110. doi:10.1080/08830180801911743. PMID 18437601.
36. See DM, Tilles JG (1996). "alpha-Interferon treatment of patients with chronic fatigue syndrome". Immunol. Invest. 25 (1–2): 153–64. doi:10.3109/08820139609059298. PMID 8675231.
37. Ojo-Amaize EA, Conley EJ, Peter JB (January 1994). "Decreased natural killer cell activity is associated with severity of chronic fatigue immune dysfunction syndrome". Clin. Infect. Dis. 18 Suppl 1: S157–9. doi:10.1093/clinids/18.Supplement_1.S157. PMID 8148445.
38. Kida K, Isozumi R, Ito M (December 2000). "Killing of human Herpes virus 6-infected cells by lymphocytes cultured with interleukin-2 or -12". Pediatr Int 42 (6): 631–6. doi:10.1046/j.1442-200x.2000.01315.x. PMID 11192519.
39.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, Zheng Z, Nahvi A, de Vries CR, Rogers-Freezer LJ, Mavroukakis SA, Rosenberg SA (October 2006). "Cancer regression in patients after transfer of genetically engineered lymphocytes". Science 314 (5796): 126–9. doi:10.1126/science.1129003. PMC 2267026. PMID 16946036.
40. Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, Jungbluth A, Gnjatic S, Thompson JA, Yee C (June 2008). "Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1". N. Engl. J. Med. 358 (25): 2698–703. doi:10.1056/NEJMoa0800251. PMC 3277288. PMID 18565862.
41. "2008 Symposium Program & Speakers". Cancer Research Institute.
42. http://www.telegraph.co.uk/earth/main.jhtml?xml=/earth/2008/06/18/scicanc118.xml[full citation needed]
43. Rotrosen D, Matthews JB, Bluestone JA (July 2002). "The immune tolerance network: a new paradigm for developing tolerance-inducing therapies". The Journal of Allergy and Clinical Immunology 110 (1): 17–23. doi:10.1067/mai.2002.124258. PMID 12110811.
44. Durham SR, Walker SM, Varga EM, Jacobson MR, O'Brien F, Noble W, Till SJ, Hamid QA, Nouri-Aria KT (August 1999). "Long-term clinical efficacy of grass-pollen immunotherapy". N. Engl. J. Med. 341 (7): 468–75. doi:10.1056/NEJM199908123410702. PMID 10441602.
45. Correale J, Farez M (February 2007). "Association between parasite infection and immune responses in multiple sclerosis". Annals of Neurology 61 (2): 97–108. doi:10.1002/ana.21067. PMID 17230481.
46. Croese J, O'neil J, Masson J, Cooke S, Melrose W, Pritchard D, Speare R (January 2006). "A proof of concept study establishing Necator americanus in Crohn's patients and reservoir donors". Gut 55 (1): 136–7. doi:10.1136/gut.2005.079129. PMC 1856386. PMID 16344586.
47. Reddy A, Fried B (January 2009). "An update on the use of helminths to treat Crohn's and other autoimmunune diseases". Parasitol. Res. 104 (2): 217–21. doi:10.1007/s00436-008-1297-5. PMID 19050918.
48. Laclotte C, Oussalah A, Rey P, Bensenane M, Pluvinage N, Chevaux JB, Trouilloud I, Serre AA, Boucekkine T, Bigard MA, Peyrin-Biroulet L (December 2008). "[Helminths and inflammatory bowel diseases]". Gastroenterol. Clin. Biol. (in French) 32 (12): 1064–74. doi:10.1016/j.gcb.2008.04.030. PMID 18619749.
49. Zaccone P, Fehervari Z, Phillips JM, Dunne DW, Cooke A (October 2006). "Parasitic worms and inflammatory diseases". Parasite Immunol. 28 (10): 515–23. doi:10.1111/j.1365-3024.2006.00879.x. PMC 1618732. PMID 16965287.
50. Brooker S, Bethony J, Hotez PJ (2004). "Advances in Parasitology Volume 58". Advances in parasitology. Advances in Parasitology 58: 197–288. doi:10.1016/S0065-308X(04)58004-1. ISBN 9780120317585. PMC 2268732. PMID 15603764.  
51. Fujiwara RT, Cançado GG, Freitas PA, Santiago HC, Massara CL, Dos Santos Carvalho O, Corrêa-Oliveira R, Geiger SM, Bethony J (2009). Yazdanbakhsh, Maria, ed. "Necator americanus infection: a possible cause of altered dendritic cell differentiation and eosinophil profile in chronically infected individuals". PLoS Negl Trop Dis 3 (3): e399. doi:10.1371/journal.pntd.0000399. PMC 2654967. PMID 19308259.
52. Carvalho L, Sun J, Kane C, Marshall F, Krawczyk C, Pearce EJ (January 2009). "Review series on helminths, immune modulation and the hygiene hypothesis: mechanisms underlying helminth modulation of dendritic cell function". Immunology 126 (1): 28–34. doi:10.1111/j.1365-2567.2008.03008.x. PMC 2632707. PMID 19120496.
53. Fumagalli M, Pozzoli U, Cagliani R, Comi GP, Riva S, Clerici M, Bresolin N, Sironi M (June 2009). "Parasites represent a major selective force for interleukin genes and shape the genetic predisposition to autoimmune conditions". J. Exp. Med. 206 (6): 1395–408. doi:10.1084/jem.20082779. PMC 2715056. PMID 19468064.


What can INFEPERIUM™’s breakthrough immunomodulators and biological response modifiers do? And why are they different? 
INFEPERIUM™’s active ingredient is one of the few molecules that affects and modulates both the innate and adaptive immune systems without negative side effects. 

INFEPERIUM™’’s immunomodulators and biological response modifiers can therapeutically treat the following conditions and more:

  • Chronic Plaque Psoriasis
  • Recalcitrant Diabetic Wounds
  • Primary Progressive MS (Multiple Sclerosis) and other Demyelinating Neuropathies and other Neurological conditions
  • Chronic Pain
  • Chronic AutoImmune Diseases
  • Various Hard-Treat Cancers

Actions Addressed by INFEPERIUM™’s immunomodulators and biological response modifiers
I. Inflammation

  • Preserving Acute inflammation which is necessary for healing, restoration and defense against invading pathogens.
  • Reducing Chronic inflammation which is the beginning of the reversal of degenerative disease.

II. Restores proper Cell Signaling

  • Through cytokines, cell messenger pathways, cellular communication and transduction.
  • Reverses aberrant, dysregulated, disrupted, error altered, confused and distracted communication.


III. Acts as an Adjuvants or Adjunct 

  • Help regulate and restore the proper on/off switches of immune cascade proteins and cell activity, especially Apoptosis (programmed cell death).
  • This will amplified proper cell activity, messaging and signal routing

The Adaptive Immune System

Adaptive immunity is a person's defense system built on specific cellular targeting. It takes time for the immune system to develop its weaponry (up to 96 hours after infection), but ultimately the adaptive response is far more effective because of its precision.

Once infection is identified, antigen is transported to lymphoid organs where it is recognized by naive B and T cells. Clonal expansion and differentiation of these cells occurs, and then the battle begins. The immune system can take several tacks, depending on the type of infection encountered. Ultimately, the goals of the adaptive response are two-fold: to produce neutralizing antibody, and to flag up infected cells for destruction. This annihilation can be carried out by the cells of both innate and adaptive immunity.

Actions of the Adaptive Immune System
Adaptive immunity is stimulated by the generic actions of innate immunity. Once a foreign organism is identified by the innate immune system, circulating T-cells begin interacting with foreign antigen. Based on their encounter, they can do one of three things: they can kill infected cells directly, they can boost the actions of macrophages to kill infected cells, or they can return to lymph tissue to incite a B cell response. 

Stimulated B cells will proceed to produce antibody, which can then circulate to fight the infection.

Antigen Recognition
"Antigen" refers to the parts of a foreign organism recognizable by the adaptive immune system. Typically, these are structural proteins, such as the spike proteins of viruses. Antigens can be huge, and are more often identified by epitopes, or smaller fragments of the folded proteins. As such, a single antigen can be recognized by multiple antigen receptors. Antibody has evolved to recognize a dizzying array of antigen epitopes. Antigen can be picked up by lymphocytes in the lymph tissues (T cells and B cells) or the blood stream (T cells only).


T cell receptors (TCR) recognize antigen fragments (that is digestion products) on the surface of cells, whereas B cell receptors (BCR) bind whole antigen in the extracellular fluid. T cells only "see" antigen when it is presented by MHC (Major Histocompatibility Complex) on the cell surface. Antigen digestion and presentation is one of the major functions of the dendritic cells (circulating monocytes) and macrophages. These are referred to as Antigen-Presenting Cells (APCs).

Naive B-cells express IgD and IgM on their cell surfaces, which bind antigen as it is washed into lymph tissue with the afferent lymph fluid. Antigen is presented to B cells by follicular dendritic cells (FDCs), which are also classed as APCs. FDCs can endocytose antigen directly from the afferent lymph or receive them from CD4+ T-cells.


Cellular response: Proliferation and Differentiation
    •    T cell response
Once T cells recognize antigen presence in the tissues, they go into action. Their first response is always to recruit help, which is accomplished by returning to the nearest lymph node to carry out clonal expansion. Daughter T cells are created with identical TCRs in order to recognize the identified antigen. These daughter cells are then returned to the circulation via the efferent lymph.


T cells can differentiate three different ways, based on their Cluster of Differentiation (CD) number. All T cells are CD3+, and naive circulating T cells will differentiate upon interaction with antigen to become either CD8+ (cytotoxic) or CD4+ (helper) T cells. CD4+ T-cells will initially become CD4-TH0 cells, and must differentiate to TH1 or TH2 depending on the whim of the adaptive response. TH1 and TH2 cells carry out different types of responses: TH1 is responsible for enhancing the macrophage response, whereas TH2 cells enhance the B cell antibody production. Typically, animals produce a balanced response of TH1 and TH2 cells, though this can lead to pathology, as can a skewed response, depending on the nature of the foreign organism.


    •    B cell response
Naive B cells recognize antigen in the lymph tissue when it is presented to them by Follicular Dendritic Cells (FDCs). They also undergo clonal expansion, creating a germinal center in the follicle as they develop and mature into plasma cells. Once mature, plasma cells in the lymph node migrate to the medullary cords and begin secreting antibody into the efferent lymph. Antibody eventually reaches the circulation in order to wage war on the intruder.


Tools of the Adaptive Immune System
Antigen Presenting Cells
    •    Macrophages
    •    Interdigitating Dendritic Cells
    ◦    Only IDCs can incite a primary response in naive T-cells
    •    CD4+ Tcells
    •    B-cells
Antigen Binding Molecules
    •    Immunoglobulins
    •    T-cell Receptor (TCR)
    •    Natural Killer (NK) Cells


Adaptive Immunity to Viruses
Humoral
    •    Production of neutralizing antibody
    •    Antibody-dependent cell mediated cytotoxicity (ADCC)
Antibody-labelled cells can be targeted by NK Cells as another defense against viral infection. Antibody produced against viral protein can attach to infected cells during their budding phase, which effectively labels them for NK targeting. NK cells express Fcγ receptors with which to detect such cells. Once activated, they release a host of enzymes to induce apoptosis of the budding cell.

Cell-Mediated
    •    CD8+ T-cell mediated killing of virus infected cells
    ◦    Main cells involved in the immune response to intracellular virus infection
    ◦    Recognition of MHC I-peptide complex
    ◦    Infected cells are killed by apoptosis
    ▪    Perforin and granzymes activate the caspase cascade
    ▪    Fas-ligand triggers the Fas-mediated apoptosis pathway
    ▪    Cytotoxic cytokines (especially TNF-α & TNF-β lymphotoxin) act on TNF receptors to induce programmed cell death


Adaptive Immunity to Bacteria
    •    The adaptive and innate responses work together to destroy bacteria
    •    The adaptive response ensures the innate response is carried out efficiently
Humoral
    •    Complement activation of the classical pathway
    ◦    Production of IgM and IgG makes the complement system more efficient
Cell-Mediated
    •    Help for macrophages
    ◦    IgG production (T-helper type II cells and B cells) which improves phagocytosis by opsonisation
    ◦    Infected macrophages are rescued by T-helper type I cells when phagocytosis and digestion mechanisms fail to eliminate the pathogen

Extracellular Infection
    •    Complement and phagocytosis
    •    B cell and T helper type II cell stimulation
    •    Production of IgM which activates the classical cascade
    •    Class switching of IgM to IgG which is a good opsonin and targets bacterial Fcγ receptor expressed by macrophages and neutrophils

Vesicular Infection
    •    The infected macrophage secretes IL-12
    •    IL-12 stimulates T-helper type I cells which release IFN-γ
    •    IFN-γ triggers the macrophages to kill the pathogens inside


The Innate Immune System
The innate immune system is the first barrier of defense to infection. It relies on an older, more generic, and faster acting set of tools than the adaptive system. While the adaptive system is essential for a specific response to infection, it is ultimately the innate system that conquers foreign attackers through means of phagocytosis.
Non-specific protective mechanisms include such innate factors as:
    •    Physical barriers
    ◦    Skin
    ◦    Ciliated mucous membranes
    ◦    Commensal organisms
    •    Humoral factors
    ◦    Lysozyme
    ◦    Complement
    ◦    Interferons
    •    Cellular mechanisms
    ◦    Phagocytosis
    •    Factors which regulate species specificity
    ◦    Membrane receptors for pathogens
    ◦    Nutritional requirements
    ◦    Temperature
    ◦    pH
    •    Mechanisms of innate immunity are always present and generally unchanging
    •    Adaptive immunity is acquired only on contact with the infectious agent (antigen) and therefore does not function before first contact with the antigen


Actions of the Innate Immune System
Recognition of Microorganisms
    •    The innate immune system recognizes components of pathogens which are intrinsically foreign (i.e. not present on normal mammalian cells), such as:
    ◦    Lipopolysaccharides of gram-negative bacteria
    ◦    Peptidoglycans of gram-positive bacteria
    ◦    Mannose sugars
    ◦    D-isoform amino acids
    •    These are given away as foreign by expressing pathogen-associated molecular patterns (PAMPs)
    •    PAMPs are recognized by pattern recognition receptors (PRRs) expressed on mammalian cells
    ◦    Pattern recognition receptors are expressed on many different cell types, not just on phagocytes
    ◦    Not all are expressed by all cells: different cell types express a different range of PRRs
    ◦    PRRs are either intracellular, membrane-associated or soluble:
    ▪    Recognition of pathogens via the cellular PRRs results in phagocytosis and inflammation
    ▪    Recognition of pathogens via the humoral PRRs results in various killing mechanisms
    •    Engagement of PRRs by PAMPs triggers:
    ◦    Phagocytosis
    ◦    The expression of cytokines, which brings about inflammation and other immune responses

Contraindications
Infeperium’s immunomodulators and biological response modifiers work as a positive adjunct for antibiotic, antiviral, and chemotherapeutic therapies. However, Corticosteroids and any other steroid therapy will immediately cause the biologic modification and immunomodulating effectiveness to cease. Therefore, all steroid therapies must cease 30 days prior to the start of any Infeperium’s immunomodulators and biological response modifiers protocol. There are no other known pharmaceutical contradictions experienced in over 10 years.

Safety Study for INFEPERIUM™’s immunomodulators and biological response modifiers

Brief description


INFEPERIUM™’s immunomodulators and biological response modifiers are presently made according to the most rigid standards of Current Good Manufacturing Practices (cGMP). The active ingredient in Infeperium’s immunomodulators and biological response modifiers has already met the rigorous standards for Veterinary use and it is approved for animal prescription under the USDA!
 
We believe INFEPERIUM™’s BRM’s to be a new category of drug that may be characterized as a Biological Response Modifier and an “Immunomodulator.” We believe that many disease pathologies that affect individuals are the result of an over-active immune system. Specifically, when a viral agent begins to adversely affect an individual’s cells, the immune system frequently becomes overactive, which destroys the viral agent but also injures surrounding healthy cell structures. We believe other disease pathologies suppress an individual’s immune system, which allows other diseases and agents to kill healthy cells. Although research is always ongoing, the product in its present form is complete. Research has concluded that Infeperium’s immunomodulators and biological response modifiers regulate the bodies immune system to prevent it from both over-reacting and under-reacting to a viral invasion of an individual’s body systems. We believe that Infeperium’s immunomodulators and biological response modifiers contains a number of unique peptide or lipopeptide molecules which may neutralize viral pathogens and their inhibitory properties by activation of a cytokine system. This, in turn, will enhance an individual’s cell mediated immunity and augment the individual’s humoral immune system possibly by eliminating negative inhibitory cytokine factors and pathogenic free-floating organisms, while simultaneously sparing normal and healthy cells.

Our objective is the continued development of Infeperium’s immunomodulators and biological response modifiers and there cognates and variants for treatment of multiple medical conditions.


Safety and Toxicity

INFEPERIUM™’s immunomodulators and biological response modifiers were not pyrogenic (endotoxin < 0.08 EU/mg).

They (≤ 1mg/mL) did not affect cell viability or inhibit protein translation as assessed in human diploid fibroblasts,

They (≤ 1mg/mL) did not induce the hemolysis of red blood cells, and

They did not promote a “Delayed Type Hypersensitivity Reaction”:  wheals did not form when 0.1 mL they (20mg/mL) was introduced by intradermal injection into New Zealand White rabbits previously exposed to Infeperium’s immunomodulators and biological response modifiers at a dose ten times a therapeutic amount.

Safety Studies with the Active Ingredient
INFEPERIUM™’s immunomodulators and biological response modifiers
Pyrogenicity of INFEPERIUM™. Pyrogenicity. The standardized endotoxic activity for two independent preparations of Infeperium was determined by Associates of Cape Cod (Woods Hole, MA) using an LAL gel clot assay (Test Date 12/6/2000 - see Unit 2B). The threshold sensitivity of the reaction was 0.03 endotoxic unit (EU) per ml. Two separate preparations of Infeperium were analyzed for endotoxic activity. Each preparation had equivalent endotoxin levels of 0.08 EU/mg. Therefore Infeperium does not contain a level of endotoxin sufficient to promote a pyrogenic response.

Hemolysis of Red Blood Cells was not Induced by Infeperium’s immunomodulators and biological response modifiers
The BRM-induced hemolytic activity in a 5% suspension of human red blood cells was measured by reading the absorbance of the hemolysate at 415 nm as described by Yoshida et al. (1994).

Study performed: The toxicity of Infeperium was tested by determining its ability to lyse human erythrocytes. Freshly collected blood (~4ml) was placed in a 15ml centrifuge tube and PBS added to bring the volume to 10ml. After centrifuging for 2-3 minutes at 2000g, the sample contained three layers: bottom layer of RBC, middle interphase of WBC, and the top supernatant layer. The supernatant and interphase layers were removed and the volume brought to 10mls and centrifuged and washed as above four more times. PBS was then added to the RBC’s to make approximately a 10% (v/v) suspension. Infeperium concentrates ranging from 1ug/ml to 1mg/ml were incubated with a 5% suspension of red blood cells and incubated for 30 minutes. Absorbance was monitored at 400 nm and hemolysis determined by comparison to control the wells that had undergone complete hemolysis by the addition of 0.2% Triton X-100 as a positive control.

The values obtained from the hemolysis assay are presented in Table 1.

None of the concentrates of INFEPERIUM™’s immunomodulators and biological response modifiers exhibited any degree of red blood cell hemolysis

Assessment of Cell Viability and Inhibition of Protein Translation in the Presence of INFEPERIUM™’s immunomodulators and biological response modifiers

Cell Viability/Fibroblast Toxicity. Infeperium isolated from caprine serum (Lot No. 111203) was used as the test article. The test article was prepared as a 1 mg/ml stock solution in media and was serially diluted in a decade series to give 1, 10, 100, and 1000 ug/ml test solutions. Test solutions thus prepared were used within one hour.

Infeperium’s immunomodulators and biological response modifiers-induced damage and inhibition of cellular replication in normal fibroblast and epithelial cell lines was monitored and found negative.


Cell Viability Assay: Human diploid fibroblasts cultures were established from skin tissue obtained from O’Bleness Medical Center, Athens, OH. Cells were maintained in Dulbecco’s modified Eagle’s media (DMEM). DMEM was prepared with L-glutamine, pyridoxine hydrochloride and without sodium pyruvate and sodium bicarbonate (GibcoR, Invitrogen, Carlsbad, CA) supplemented to a final concentration of 100 units/ml of penicillin and 100 micrograms/ml of streptomycin (penicillin-streptomycin, GibcoR, Invitrogen, Carlsbad, CA), and adjusted to contain 10% NuSerum I (Collaborative Research Products, Bedford, MA).


The cells were transferred into 12-well culture plates at a concentration of 5X104 cells/cm2 (in 2 ml media) and incubated for 24 hours at 37 degrees C with 5% CO2. After the cells were incubated for 36 hours, the media was removed and replaced with 2ml of the test article Infeperium at concentrations from 1ug/ml to 1mg/ml dissolved in DMEM media. Three wells at each concentration of test article were prepared on two separate plates to provide triplicate measurements at 24 hours. A control plate was established with three wells of cells to which DMEM media (2ml) only was added in place of the test article. The control plate was incubated as described above. Three control wells were read at 24 hours.


When the test or control plate was removed from the incubator (24 hours), the supernatant was aspirated with 50ul of 0.05% trypsin/EDTA (Invitrogen) added to each well. The plate was incubated at 37 C for two hours. Then, 950ul fresh DMEM was added to each well and cells responded. After mixing to ensure homogeneity, the cells were counted by the trypan blue dye exclusion method: to each 100ul of cell volume, 100ul of 0.4% trypan blue is added. After mixing, the hemocytometer was loaded and the number of live cells (those that had excluded trypan blue) counted in the four counting fields. The number of cell/ml was calculated by the following formula: Cells/ml=average of total cells counted *dilution factor* volume hemocytometer.


Results: Incubation of cells with concentrates of Infeperium’s immunomodulators and biological response modifiers up to 1 mg/ml did not decrease cell viability.

A similar test was performed by Viromed, as part of the Infeperium’s immunomodulators and biological response modifiers assessment study for which they were contracted. Using the A-72 canine fibroblast cell line they determined that Infeperium was not cytotoxic (cellular proliferation was not impaired). These results can be found in the Viromed documentation provided (project numbers: 10295 and 10296, following the assigned protocol numbers: MS121200-AV and MS121200-V).

Translation Inhibition Assay: After fibroblast cells were incubated for 24 hours as described above, the media was aspirated and the cells washed twice with fresh met-cya-media. The test article at concentrations of 0.001, 0.01, 0.1, and 1 mg/ml in [35S]met-cys-media (GibcoR, Invitrogen, Carlsbad, CA) to give a final specific activity of 20uCi/ml were prepared. Each test article concentration (0.5ml for 1 hr, 1.0ml for 24 hr) was added to each of three wells (n=3) on each of two culture plates as described above. The plates were incubated for either 1 hour or 24 hours. After incubation, the media from each of the wells was discarded and the wells washed twice with PBS. Trichloroacetic acid (0.5ml) was added to each well and the cells removed with a rubber policeman, transferred to microcentrifuge tubes, and incubated for 30 minutes on ice. The incorporation of [35S]methionine was measured by liquid scintillation counting.


Results: At 1 hour after addition of Infeperium’s immunomodulators and biological response modifiers, significant translation inhibition was only noted at a test concentration of 1mg/ml. However 24 hours after addition of Infeperium’s immunomodulators and biological response modifiers, translation inhibition was not noted at any of the test concentrations of Infeperium’s immunomodulators and biological response modifiers. At 1 hour after addition of test sample, the high concentration of Infeperium’s immunomodulators and biological response modifiers may have swamped the ability of the cell to incorporate radiolabel. However, as noted at 24 hours, equilibrium was established over time, thus demonstrating that Infeperium’s immunomodulators and biological response modifiers has no effect on protein translation over time.


Discussion: Cell viability was chosen as a study parameter as it provides an easily visualized and understood answer for a drug’s toxicity to a cell line. Since cell viability can show a degree of variance, an additional parameter was chosen to be assessed—inhibition of protein translation. Using this method we would be able to detect subtle, sub-lethal changes in cell viability. By performing both of these studies, we were able to demonstrate that Infeperium’s immunomodulators and biological response modifiers has no effect on cell viability or on translation of human fibroblast cells.

Delayed Type Hypersensitivity
Delayed Type Hypersensitivity. Six New Zealand White rabbits (3 animals per group) received either 5mg of Infeperium in 0.25ml Freund’s Complete Adjuvant (FCA) or 0.25ml FCA by subcutaneous injection. On days 4, 11, and 21 one rabbit from each group was selected to receive an intradermal injection of 0.1ml (200ug) of Infeperium and sterile water (negative control) on the exposed lateral thorax. After 15-30 minutes the injections sites were observed for raised erythematous lesions (wheals) at the injection site. No wheal development was observed indicating the absence of a delayed type hypersensitivity reaction to Infeperium’s immunomodulators and biological response modifiers.

Safety Testing of USDA approved product which contains the same active ingredient of Infeperium’s immunomodulators and biological response modifiers.
Over 100,000 animals were tested, species ranging from horses, pigs, sheep and cows, without any negative events recorded including no systemic anaphylactic reactions.

A range finding safety study in rats and guinea pigs was performed lasting two weeks after dosage, the following tissues were examined and submitted for histopathologic examinations.

The following tissues were taken at necropsy:

Gross Pathology Results
No gross lesions were seen.

Histopathology Results
No lesions considered to be treatment related were seen.

Discussion and Conclusions
Three doses were chosen for this study—half (0.75ml), double (3.0ml), and the standard human clinical dose (1.5ml) into rats and guinea pigs.

By contrast, a 60kg human receiving a standard clinical dose of Infeperium (1.5ml) would be receiving a dose of 0.025ml Infeperium/kg, a dose 700 times lower than the top dose used in rats and guinea pigs in this study. This indicates a potentially very favorable safety profile for this agent.

Clinical Assessment of Safety
From the report dated September 5, 2005, I was directly involved in assessing the safety and efficacy of a novel biological product. This involved the treatment of patients on an informed consent basis.

During the aforementioned period, I was personally responsible for the treatment and clinical management of approximately 200 patients. The greater majority of whom, received the ‘Product’ for the entire duration of that time period.

During that time clinical data was collated by my own observations as well as from patients’ diaries which were self-recorded on a daily basis.  

The collected observational, clinical and scientific data was subsequently presented to the Ethics Committee and MHRA (Medical Health Regulatory Authority) in order to obtain permission for full human clinical trials. This permission was granted on the basis that the ‘Product’ appeared to be safe and efficacious.

Treatment Regimen
The ‘Product’ was administered by sub-cutaneous injection in variable dosages, according to response.

Initially a test dose of 0.1mls was administered. The reason for the test dose was to ascertain that there was no allergic response or in its severest form, anaphylactic shock.

Thirty minutes after administration of the test dose a further 0.9ml was given again by sub-cutaneous injection. Patients remained in the waiting area for an additional 30 minutes, in order to ascertain that there were no adverse side-effects which required recording and/or addressing.

Subsequently all patients attended weekly or bi-weekly for treatment. The dose given varied from 1ml to 4mls - according to the clinical response of the patient.

All patients were instructed to keep a daily diary of their condition, either written or taped.

Clinician’s Summary
Having worked closely with approximately 200 patients over a period of 30 months, I am in a unique position to state that I am extremely impressed with both the benefits and safety of this product.

Firstly, the product itself is extremely well tolerated, except for a reasonable proportion of patients who have a mild localized reaction at the site of injection, this reaction is akin to an “insect bite”, and attenuated by the use of antihistamines.

Secondly, in two and a half years, no patient has exhibited any adverse reactions of note. The apparent lack of side effects is quite amazing and totally unexpected. This is supported by the observations of the two clinical trials that were undertaken in the UK.

In a number of patient’s blood samples were taken for routine testing, and in all cases the parameters were entirely normal (Renal Function, Liver Function, Full blood Count, etc.).
 
It must be noted that all the above held true across the dosage range of 1ml to 8ml per week.

Conclusion
It is my professional opinion that this novel biological product appears to be a safe and effective, beneficial treatment and free from major side effects.
To date no individual has reported any side effect of note.

Disclaimer

This Webpage is for general health information only. This page is not to be used as a substitute for medical advice, diagnosis or treatment of any health condition or problem. Users of this Webpage should not rely on information provided on this page for their own health problems. Any questions regarding your own health should be addressed to your own physician or other healthcare provider.

No one associated with this Webpage makes any warranties nor express or implied representations whatsoever regarding the accuracy, completeness, timeliness, comparative or controversial nature, or usefulness of any information contained or referenced on this Webpage. Nor do we assume any risk whatsoever for your use of this webpage or the information contained herein. Health-related information changes frequently and therefore information contained on this Web site may be outdated, incomplete or incorrect. Statements made about products have not been evaluated by the Food and Drug Administration. Use of this Webpage does not create an expressed or implied physician-patient relationship.

This Webpage does not record specific website user information and does not contact users of this website. You are hereby advised to consult with a physician or other professional health-care provider prior to making any decisions, or undertaking any actions or not undertaking any actions related to any health care problem or issue you might have at any time, now or in the future. In using this website you agree that no party affiliated with this Webpage, nor any other party is or will be liable or otherwise responsible for any decision made or any action taken or any action not taken due to your use of any information presented at this website.

WARNING!!! This page is for information purposes only. It is intended for the use of licensed physicians and medical researchers. If you do not qualify, please leave this page. The FDA has neither approved or disapproved comments made on this website. The exception would be direct quotes and references made by an agency of the U.S. government, such as the FDA, CDC or NIH.

>