Neuro

Open journal

ISSN 2377-1607

Current Utilization of Mast Cell Stabilizers for Preemptive Treatment of NF1 Neurofibromas

Vincent M. Riccardi*

Vincent M. Riccardi, MD, MBA

The Neurofibromatosis Institute, 5415 Briggs Avenue, La Crescenta, CA 91214, USA; Tel. 818-957-3508; E-mail: Riccardi@medconsumer.com

INTRODUCTION

The morbidity and mortality of NF1 are directly and indirectly related to the person’s neurofibroma burden. The person with more and/or larger neurofibromas is at higher risk. Most persons with NF1 are born absent neurofibromas of any type. However, I estimate that between 5% and 15% of NF1 persons have one or more congenital neurofibromas. These are properly referred to as Diffuse Plexiform Neurofibromas (DPN)1,2 and more technically as epineurial neurofibromas.1 In addition, almost all NF1 persons are also at risk for the later development of cutaneous neurofibromas (endoneurial neurofibromas)1 and some NF1 persons are at risk for subcutaneous and nodular plexiform neurofibromas (both of which are perineurial neurofibromas).1 The latter are likely associated with higher mortality3,4 and they are especially common in persons with NF1 on the basis of a “whole gene deletion.”5,6

For the most part, the routine approach to NF1 has been to wait for progression of the NF1 neurofibroma burdens to have major, serious clinical consequences and then use very expensive medications that often have serious side-effects and uncertain abilities to stop or reverse the neurofibroma’s progression. In this presentation, I suggest greater reliance on preemption – minimize the initiation and progression of these mass lesions.

The NF1 neurofibromas can have direct and indirect adverse consequences, precisely as neurofibromas, depending on absolute size, adjacent structures and infiltration into surrounding tissue. Endoneurial (cutaneous) neurofibromas usually first appear in the teenage years, ultimately often accounting for hundreds, even thousands of individual lesions, varying in size from a few grams7 to several kilograms.7 At any size they are almost always cosmetically compromising and are the sites of localized itching, pain and tenderness. Perineurial (subcutaneous and nodular plexiform) neurofibromas may occur at any age and are often a source of substantial pain and focal neurological deficits. Nodular plexiform neurofibromas are often paraspinal at multiple sites.1,8,9 In addition, a minimum 10% of NF1 patients will experience malignant transformation of one or more plexiform neurofibromas, perineurial or epineurial (DPN), the malignant tumor usually a sarcoma, most often a neurofibrosarcoma.10,11,12 While it has not been proven that a mast cell blocker preemption approach decreases the malignant transformation risk, it seems likely – even compelling – that an arrested neurofibroma is less likely to realize this transformation. Can there be a better reason for attempting to arrest the growth of NF1 neurofibromas? 

REVIEW

Pathologists have known for decades that mast cells are an intrinsic element of neurofibromas, whether or not NF1 is present.13,14,15,16,17,18,19,20,21 In some instances, NF1 neurofibroma mast cells have been quantified and there is consistently an excess compared to normal tissue.22,23 However, no attempts seem to have been made to identify the tissue source or detailed nature of the NF1 neurofibroma’s mature mast cells, for example, tryptase or chymase predominance.24,25 Likewise, no efforts have been made to distinguish the immature mast cell26,27 newly arrived to the incipient neurofibroma in contrast to the “mature,” more or less neurofibroma-specific mast cell. Targeting the immature mast cell may be a key to arresting the incipient or very early stage neurofibroma.28 In any event, numerous pathologists established a role for mast cells in neurofibroma pathogenesis.

Respecting both the pathology data just noted and consistent with NF1 patient reports that likely sites of neurofibroma development were heralded by focal intense itching,12,29,30,31 a symptom well-known to indicate mast cell infiltration, Riccardi began emphasizing a key role for mast cells and cell-cell interactions in NF1 neurofibroma pathogenesis.30,32 A key part of the proffered schema was consideration that the early stages of the neurofibroma are more like wound-healing dysplasia than neoplasia sensu strictu.7,33,34

Most early (and even many present-day) neurofibroma investigators presumed automatically that a neurofibroma is a tumor from the outset, with a somatic mutation in the normal NF1 allele (second hit) in a Schwann cell being the literal “cause” of the NF1 neurofibroma.35,36,37,38 This myopic approach ignores the cogent possibility that the initial dynamics of NF1 neurofibroma formation were more related to wound-healing, the wound providing a cellular and extra-cellular milieu affording an increased likelihood of the second hit (incubator effect).8 Trauma (mechanical, biochemical, hypoxic) and wound-healing became a focus of NF1 neurofibroma pathogenesis in the very early 1980’s.39,40,41,42,43,44,45,46

Along the way, in 1988, Giorno, et al.,47 documented that human NF1 neurofibroma fibroblasts (a major portion of the lesion) incorporate mast cell granules into their cytoplasm, consistent with the work of others.48 These results were also consistent with the massive data generated by the Wade Clapp University of Indiana group using Nf1 mutant mice.4960 They professed and nurtured the notion that “mast cells are necessary, but not sufficient” to generate Nf1+/- neurofibromas. One published commentary on that work noted the compatibility of this mouse model with the Riccardi approach.51 And a 2009 publication61 showed that the mast cell blocker, tranilast, impaired the ability of Nf1+/- mice neurofibroma to progress.

Further indication that mechanical trauma initiated mouse Nf1+/- neurofibromas was established by A.C. Lloyd’s group.62 Wound-healing was also emphasized by others.41,45,63,64,65 The Lloyd group’s article just cited also established that normal (Nf1+/+) mast cells were as efficient as Nf1+/- mast cells in generating and sustaining mouse Nf1+/- neurofibromas. In other words, it is likely that the mast cell per se and its ordinary functions contribute to human NF1 neurofibroma initiation and progression. The NF1 mast cell as a therapeutic target of drugs aimed at normal mast cells, as, for example, in asthma and other mast-cell- associated disorders, was and is thereby even more compelling.

All of these developments were in the context of Riccardi’s publications documenting that ketotifen (Zaditen®), in both open-label and double-blind protocols, had obvious benefits in treating NF1 neurofibromas. As early as 1987, such benefits were published in the Archives of Dermatology.66 Additional data were published in 1990 and 1993.33,67 In the 20-plus years since their publication, these data and the conclusions derived therefrom have been casually challenged once,68,69 but never refuted or contradicted. The published benefits of NF1 ketotifen treatment included 1) a decrease in the itching, pain and tenderness associated with neurofibromas of all three types, especially for the endoneurial (cutaneous) neurofibromas and subcutaneous perineurial neurofibromas; 2) a decrease in the rate of appearance of cutaneous neurofibromas; 3) a decrease in the rate of enlargement (“growth”) of all types of neurofibromas; 4) a major decrease in intra-operative small-vessel hemorrhage in plexiform neurofibromas; and 5) a consistent declaration of improvement in the NF1 person’s overall sense of well-being. Parenthetically, this latter phenomenon has been noted in patients treated with ketotifen to minimize surgical scars.70 In 1998, for three German NF1 patients were reported71 to have the same salutary results from ketotifen treatment as reported by Riccardi.

In 2015, I reported the results of 30 years of ketotifen treatment of an NF1 patient from age three months.72 At age 30, his skin was universally free of any mature neurofibromas. Instead, there was only the monotonous presence of “early,” small and flat cutaneous neurofibromas. It was as though these neurofibromas had been arrested and maintained in this very early phase of neurofibroma development. In addition, the patient’s right ankle DPN was much smaller than would have been expected without treatment. At about the same time, I established NFormation, an online venue to report on advances in NF1 research (www.medconsumer.com). Included were a series of American, European and South American NF1 persons who were undergoing or had undergone self-determined treatment with ketotifen or an alternative mast cell blocker. The consistency of the self-reported results was impressive, both within the self-determined treatment group and when compared with the three protocol-based reports noted above. Finally, the combined data are sufficiently robust to have instigated the already-begun preemptive treatment of a one year old child with NF1 for whom there is expectation of a high cutaneous neurofibroma burden.

Although ketotifen has not been approved by the American Food and Drug Administration (FDA) other than for eye-drops (Zaditor®), there are multiple active investigational ketotifen protocols in the USA and elsewhere to study this drug’s influence on minimizing fibrosing conditions73,74,75 and excessive scarring in surgical and other wounds.76,77,78,79,80 Ketotifen is also used extensively world-wide for asthma, atopic dermatitis, helminthic infections, and various eating disorders, among others.81,82 With such extensive clinical usage, its relative safety is well established. It is relatively inexpensive, with costs per tablet in the range of 15 cents or so. It is now available in generic form and it remains to be established that all of the available brands are equivalent to Zaditen®, the only preparation with which I have extensive personal experience. It also should be noted that the FDA’s failure to approve ketotifen was based on that agency’s presumption of poor efficacy for treating asthma. Specifically, ketotifen did not have toxicity or adverse effects that contributed to denial of FDA approval.

One of the conditions sometimes very effectively treated with ketotifen is systemic mastocytosis.25,83,84,85 This pronouncement brings up the probability that NF1 may be a form of mastocytosis, a consideration raised earlier by myself86 and others.87 Ultimately, these and many other considerations will be relevant to the long-term role of ketotifen and other mast cell blockers, for example, the identification of effective biomarkers88 and the influence of vitamin D congeners.89 However, our concerns presently are on if and how soon mast cell blockersespecially ketotifen – are made available to all persons with NF1. This issue is especially critical for Americans, given the lack of FDA approval of ketotifen, the literal and figurative fuel for elaborating the arguments propounded here. 

CONCLUSION

Ketotifen and likely similar mast cell blockers have the potential to reduce drastically the NF1 neurofibroma burden on a world-wide basis. Given the data provided herein, it is not clear why this potential has been overlooked, ignored or dismissed. I can only hope that these few words will make this treatment strategy immediately available to at least some of the earth’s estimated two million or more persons alive with NF1 today.

CONFLICTS OF INTEREST

I, the sole author of this manuscript, have no conflicts of interest to declare. Specifically, I have no financial interests of any sort, direct or indirect, with any for-profit organization or semblance thereto. I will realize no financial or other compensation consequent to or otherwise associated with this manuscript.

ACKNOWLEDGMENTS

Thanks to the many NF1 patients and their families for their participation in the formal and informal protocols and their cooperation in finalizing the data. “The Neurofibromatosis Institute” is simply an alternative name for my clinical, research and publication activities; it has no legal status.

1. Riccardi VM. The genetic predisposition to and histogenesis of neurofibromas and neurofibrosarcoma in neurofibromatosis type 1. Neurosurg Focus. 2007; 22(6): E3. doi: 10.3171/foc.2007.22.6.4

2. Sehgal VN, Srivastava G, Aggarwal AK, Oberai R. Plexiform neurofibromas in neurofibromatosis type 1. Int J Dermatol. 2009; 48: 971-974. doi: 10.1111/j.1365-4632.2008.04041.x

3. Pasmant E, Sabbagh A, Spurlock G, et al. NF1 microdeletions in neurofibromatosis type 1: from genotype to phenotype. Hum Mutat. 2010; 31: E1506-E1518. doi: 10.1002/humu.21271

4. Brown RM, Klesse LJ, Le LQ. Cutaneous features predict paraspinal neurofibromas in neurofibromatosis type 1. J Invest Dermatol. 2010; 130: 2167-2169. doi: 10.1038/jid.2010.206

5. De Raedt T, Brems H, Wolkenstein P, et al. Elevated risk for MPNST in NF1 microdeletion patients. Am J Hum Genet. 2003; 72: 1288-1292. doi: 10.1086/374821

6. Khosrotehrani K, Bastuj-Garin S, Riccardi VM, Birch P, Friedman JM, Wolkenstein P. Subcutaneous neurofibromas are associated with mortality in neurofibromatoss 1: a cohort study of 703 patients. Am J Med Genet A. 2005; 132: 49-53. doi: 10.1002/ajmg.a.30394

7. Riccardi VM, Eichner JE. Neurofibromatosis: phenotype, natural history, and pathogenesis. 1st ed. Baltimore: Johns Hopkins University Press; 1986.

8. Chen Z, Liu C, Patel AJ, Liao CP, Wang Y, Le LQ. Cells of origin in the embryonic nerve roots for NF1-associate plexiform neurofibromas. Cancer Cell. 2014; 26: 695-706. doi: 10.1016/j.ccell.2014.09.009

9. Carod-Artal FJ, Melo M, da Silva RT, Rizzo I, Vazquez C, Brenner C. Type I neurofibromatosis presenting as a progressive cervical myelopathy. The first case reported in Kaxinawa Indians. Rev Neurol. 2000; 31: 307-310.

10. Riccardi VM, Powell PP. Neurofibrosarcoma as a complication of von Recklinghausen neurofibromatosis. Neurofibromatosis. 1989; 2: 152-165.

11. Mautner VF, Friedrich RE, Von Deimling A, et al. Malignant peripheral nerve sheath tumors in Neurofibromatosis type 1: MRI supports the diagnosis of malignant plexiform neurofibroma. Neuroradiology. 2003; 45: 618-625. doi: 10.1007/s00234-003-0964-6

12. Riccardi VM. Neurofibromatosis: phenotype, natural history and pathogenesis. 2 ed. Baltimore: Johns Hopkins University Press; 1992.

13. Lascano EF. Mast cells in human tumors. Cancer. 1958; 6: 1110-1113. doi: 10.1002/1097-0142(195811/12)11:6<1110::aid-cncr2820110604>3.0.co;2-g

14. Cawley EP, Hoch-Ligitti C. Association of tissue mast cells and skin tumors. Arch Dermatol. 1961; 83: 92-96. doi: 10.1001/archderm.1961.01580070098010

15. Pineda A. Mast cells-their presence and ultrastructural characteristics in peripheral nerve tumors. Arch Neurol. 1965; 13: 372-382. doi: 10.1001/archneur.1965.00470040038006

16. Issacson P. Mast cells in benign nerve sheath tumors. J Pathol. 1976; 119: 193-196. doi: 10.1002/path.1711190402

17. Reed ML, Jacoby RA. Cutaneous neuroanatomy and neuropathology. Normal nerves, neural-crest derivatives, and benign neural neoplasms in the skin. Am J Dermatopathol. 1983; 5: 335-362.

18. Kirkpatrick CJ, Curry A. Interaction between mast cells and perineurial fibroblasts in neurofibromas. Pathol Res Pract. 1988; 183: 453-458. doi: 10.1016/S0344-0338(88)80092-X

19. Johnson MD, Kamso-Pratt J, Federspiel CF, Whetsell WO, Jr. Mast cell and lymphoreticular infiltrates in neurofibromas. Comparison with nerve sheath tumors. Arch Pathol Lab Med. 1989; 113: 1263-1270.

20. Donhuijsen K, Sastry M, Volker B, Leder LD. Mast cell frequency in soft tissue tumors. Relation to type and grade of malignancy. Pathol Res Pract. 1992; 188: 61-66. doi: 10.1016/S0344-0338(11)81157-X

21. Sanguinetti C, Greco F, De Palma L, Specchia N, Toesca A, Nori S. The ultrastructure of peripheral neurofibroma: the role of mast cells and their interaction with perineurial cells. Ital J Orthop Traumatol. 1992; 18: 207-216.

22. Carr NJ, Warren AY. Mast cell numbers in melanocytic naevi and cutaneous neurofibromas. J Clin Pathol. 1993; 46: 86-87. doi: 10.1136/jcp.46.1.86

23. Nurnberger M, Moll I. Semiquantitative aspects of mast cells in normal skin and in neurofibromas of neurofibromatosis types 1 and 5. Dermatology. 1994; 188: 296-299. doi: 10.1159/000247170

24. Hermes B, Feldmann-Boddeker I, Welker P, et al. Altered expression of mast cell chymase and tryptase and of c-Kit in human cutaneous scar tissue. J Invest Dermatol. 2000; 114: 51-55. doi: 10.1046/j.1523-1747.2000.00837.x

25. Kurosawa M, Amano H, Kanbe N, et al. Heterogeneity of mast cells in mastocytosis and inhibitory effect of ketotifen and ranitidine on indolent systemic mastocytosis. J Allergy Clin Immunol. 1997; 100: S25-S32. doi: 10.1016/s0091-6749(97)70001-0

26. Dudeck A, Leist M, Rubant S, et al. Immature mast cells exhibit rolling and adhesion to endothelial cells and subsequent diapedesis triggered by E- and P-selectin, VCAM-1 and PECAM-1. Exp Dermatol. 2010; 19: 424-434. doi: 10.1111/j.1600-0625.2010.01073.x

27. Garg K, Ryan JJ, Bowlin GL. Modulation of mast cell adhesion, proliferation, and cytokine secretion on electrospun bioresorbable vascular grafts. J Biomed Mater Res A. 2011; 97: 405-413. doi: 10.1002/jbm.a.33073

28. Riccardi VM. Ketotifen suppression of NF1 neurofibroma growth over 30 years. Am J Med Genet A. 2015; 167(7): 1570-1577. doi: 10.1002/ajmg.a.37045

29. Riccardi VM. The pathophysiology of neurofibromatosis. IV. Dermatologic insights into heterogeneity and pathogenesis. J Am Acad Dermatol. 1980; 3: 157-166.

30. Riccardi VM. Cutaneous manifestations of neurofibromatosis cellular interaction, pigmentation, and mast cells. Birth Defects. 1981; 17(2): 129-145.

31. North CA, North RB, Epstein JA, Piantadosi S, Wharam MD. Low-grade cerebral astrocytomas: survival and quality of life after radiation therapy. Cancer. 1990; 66: 6-14. doi: 10.1002/1097-0142(19900701)66:1<6::AID-CNCR2820660103>3.0.CO;2-F

32. Riccardi VM. Cell-cell interaction as an epigenetic determinant in the expression of mutant neural crest cells. Birth Defects. 1979; 15(B): 89-98.

33. Riccardi VM. The potential role of trauma and mast cells in the pathogenesis of neurofibromas. In: Ishibashi Y, Hori Y, eds. Tuberous sclerosis and neurofibromatosis: epidemiology, pathophysiology, biology and management. 1st ed. Amsterdam: Elsevier; 1990: 167-190.

34. Riccardi VM. Histogenesis control genes: embryology, wound healing and NF1 (Letter to the Editor). Teratology. 2000; 62: 4. doi: 10.1002/1096-9926(200007)62:1<4::AID-TERA2>3.0.CO;2-Q

35. De Raedt T, Maertens O, Chmara M, et al. Somatic loss of wild type NF1 allele in neurofibromas: Comparison of NF1 microdeletion and non-microdeletion patients. Genes Chromosomes Cancer. 2006; 45: 893-904. doi: 10.1002/gcc.20353

36. Spurlock G, Griffiths S, Uff J, Upadhyaya M. Somatic alterations of the NF1 gene in an NF1 individual with multiple benign tumours (internal and external) and malignant tumour types. Fam Cancer. 2007; 6: 463-471. doi: 10.1007/s10689-007-9149-5

37. Spyk SL, Thomas N, Cooper DN, Upadhyaya M. Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Hum Genomics. 2011; 5: 623-690. doi: 10.1186/1479-7364-5-6-623

38. Thomas L, Kluwe L, Chuzhanova N, Mautner V, Upadhyaya M. Analysis of NF1 somatic mutations in cutaneous neurofibromas from patients with high tumor burden. Neurogenetics. 2010; 11(4): 391-400. doi: 10.1007/s10048-010-0240-y

39. Levi-Schaffer F, Kupietzky A. Mast cells enhance migration and proliferation of fibroblasts into an in vitro wound. Exp Cell Res. 1990; 188: 42-49. doi: 10.1016/0014-4827(90)90275-F

40. Koivunen J, Karvonen SL, Yla-Outinen H, Aaltonen V, Oikarinen A, Peltonen J. NF1 tumor suppressor in epidermal wound healing with special focus on wound healing in patients with type 1 neurofibromatosis. Arch Dermatol Res. 2005; 296: 547-554. doi: 10.1007/s00403-005-0564-x

41. Hebda PA, Collins MA, Tharp MD. Mast cell and myofibroblast in wound healing. Dermatol Clin. 1993; 11: 685-696.

42. Grieb G, Steffens G, Pallua N, Bernhagen J, Bucala R. Circulating fibrocytes-biology and mechanisms in wound healing and scar formation. Int Rev Cell Mol Biol. 2011; 291: 1-19. doi: 10.1016/B978-0-12-386035-4.00001-X

43. Artuc M, Hermes B, Steckelings UM, Grutzkau A, Henz BM. Mast cells and their mediators in cutaneous wound healing-active participants or innocent bystanders? Exp Dermatol. 1999; 8: 1-16. doi: 10.1111/j.1600-0625.1999.tb00342.x

44. Gallant-Behm CL, Hildebrand KA, Hart DA. The mast cell stabilizer ketotifen prevents development of excessive skin wound contraction and fibrosis in red Duroc pigs. Wound Repair Regen. 2008; 16: 226-233. doi: 10.1111/j.1524-475X.2008.00363.x

45. Atit RP, Crowe MJ, Greenbalgh DG, Wenstrup RJ, Ratner N. The Nf1 tumor suppressor regulates mouse skin wound healing, fibroblast proliferation and collagen deposited by fibroblasts. J Invest Dermatol.1999; 112: 835-842. doi: 10.1046/j.1523-1747.1999.00609.x

46. Dvorak AM, Kissell S. Granule changes of human skin mast cells characteristic of piecemeal degranulation and associated with recovery during wound healing in situ. J Leukocyte Biol. 1991; 49: 197-210. doi: 10.1002/jlb.49.2.197

47. Giorno R, Lieber J, Claman HN. Ultrastructural evidence for mast cell activation in a case of neurofibromatosis. Neurofibromatosis. 1988; 2: 35-41.

48. Jones CJ, Kirkpatrick CJ, Stoddart RW. An ultrastructure study of the morphology and lectin-binding properties of human mast cell granules. Histochem J. 1988; 183: 453-461. doi: 10.1007/bf01002429

49. Ingram DA, Yang F-C, Travers JB, et al. Genetic and biochemical evidence that haploinsufficiency of the Nf1 tumor suppressor gene modulates melanocyte and mast cell fates in vivo. J Exp Med. 2000; 191: 181-188. doi: 10.1084/jem.191.1.181

50. Yang F-C, Ingram DA, Chen S, et al. Neurofibromin-deficient Schwann cells secrete a potent migratory stimulus for Nf1+/- mast cells. J Clin Invest. 2003; 112: 1851-1861. doi: 10.1172/JCI200319195

51. Viskochil D. It takes two to tango: mast cell and Schwann cell interactions in neurofibromas. J Clin Invest. 2003; 112: 1791-1793. doi: 10.1172/JCI200320503

52. Yang FC, Ingram DA, Chen S, et al. Neurofibromin-deficient Schwann cells secrete a potent migratory stimulus for Nf1+/- mast cells. J Clin Invest. 2003; 112: 1851-1861. doi: 10.1172/jci19195

53. McDaniel AS, Allen JD, Park SJ, et al. Pak1 regulates multiple c-Kit mediated Ras-MAPK gain-in-function phenotypes in Nf1+/- mast cells. Blood. 2008; 112: 4646-4654. doi: 10.1182/blood-2008-04-155085

54. Yang FC, Ingram DA, Chen S, et al. Nf1-dependent tumors require a microenvironment containing Nf1+/– and c-kit-dependent bone marrow. Cell. 2008; 135: 437-448. doi: 10.1016/j.cell.2008.08.041

55. Reilly KM, Van Dyke T. It takes a (dysfunctional) village to raise a tumor. Cell. 2008; 135: 408-410. doi: 10.1016/j.cell.2008.10.009

56. Yang FC, Chen S, Clegg T, et al. Nf1+/- mast cells induce neurofibroma-like phenotypes through secreted TGF-beta signaling. Hum Mol Genet. 2006; 15: 2421-2437. doi: 10.1093/hmg/ddl165

57. Staser K, Yang FC, Clapp DW. Mast cells and the neurofibroma microenvironment. Blood. 2010; 116: 157-164. doi: 10.1182/blood-2009-09-242875

58. Staser K, Yang FC, Clapp DW. Plexiform neurofibroma genesis: questions of Nf1 gene dose and hyperactive mast cells. Curr Opin Hematol. 2010; 17: 287-293. doi: 10.1097/MOH.0b013e328339511b

59. Chen S, Burgin S, McDaniel A, et al. Nf1-/- Schwann cell-conditioned medium modulates mast cell degranulation by c-Kit-mediated hyperactivation of phosphatidylinositol 3-kinase. Am J Pathol. 2010; 177: 3125-3132. doi: 10.2353/ajpath.2010.100369

60. Yang FC, Staser K, Clapp DW. The plexiform neurofibroma microenvironment. Cancer Microenviron. 2012; 5: 307-310. doi: 10.1007/s12307-012-0115-x

61. Yamamoto M, Yamauchi T, Okano K, Takahashi M, Watabe S, Yamamoto Y. Tranilast, an anti-allergic drug, down-regulates the growth of cultured neurofibroma cells derived from neurofibromatosis type 1. Tohoku J Exp Med. 2009; 217: 193-201. doi: 10.1620/tjem.217.193

62. Ribeiro S, Napoli I, White IJ, et al. Injury signals cooperate with nf1 loss to relieve the tumor-suppressive environment of adult peripheral nerve. Cell Rep. 2013; 5. doi: 10.1016/j.celrep.2013.08.033

63. Grenz A, Eltzschig HK. Mast cells and intestinal injury: a novel link between hypoxia and inflammation. Crit Care Med. 2013; 41: 2246-2248. doi: 10.1097/CCM.0b013e318283cc70

64. Oskeritzian CA. Mast cells and wound healing. Adv Wound Care (New Rochelle). 2012; 1: 23-28. doi: 10.1089/wound.2011.0357

65. Enoksson M, Lyberg K, Moller-Westerberg C, Fallon PG, Nilsson G, Lunderius-Andersson C. Mast cells as sensors of cell injury through IL-33 recognition. J Immunol. 2011; 186: 2523-2528. doi: 10.4049/jimmunol.1003383

66. Riccardi VM. Mast cell stabilization to decrease neurofibroma growth: preliminary experience with ketotifen. Arch Dermatol. 1987; 123: 1011-1016. doi: 10.1001/archderm.1987.01660320053011

67. Riccardi VM. A controlled multiphase trial of ketotifen to minimize neurofibroma-associated pain and itching. Arch Dermatol. 1993; 129: 577-581. doi: 10.1001/archderm.1993.01680260047004

68. Krause L. Ketotifen and neurofibromatosis. Arch Dermatol. 1988; 124: 651-652.

69. Riccardi VM, Huston DP. Ketotifen and neurofibromatosis. Arch Dermatol. 1988; 124: 652. doi: 10.1001/archderm.1988.01670050010004

70. Hellal F, Hurtado A, Ruschel J, et al. Microtubule stabilization reduces scarring and causes axon regeneration after spinal cord injury. Science. 2011; 331: 928-931. doi: 10.1126/science.1201148

71. Haustein UF. Ketotifen inhibits urticaria and tumor progression in neurofibromatosis. Dermatol Monatsschr. 1989; 175: 581-584.

72. Anastasiadou E, Slack FJ. Cancer: malicious exosomes. Science. 2014; 346: 1459-1460. doi: 10.1126/science.aaa4024

73. Walker M, Harley R, LeRoy EC. Ketotifen prevents skin fibrosis in the tight skin mouse. J Rheumatol. 1990; 17: 57-59.

74. Qu Z, Adelson DL. Bovine ncRNAs are abundant, primarily intergenic, conserved and associated with regulatory genes. PLoS ONE. 2012; 7: e42638. doi: 10.1371/journal.pone.0042638

75. Overed-Sayer C, Rapley L, Mustelin T, Clarke DL. Are mast cells instrumental for fibrotic diseases? Front Pharmacol. 2013; 4: 174. doi: 10.3389/fphar.2013.00174

76. Monument MJ, Hart DA, Befus AD, Salo PT, Zhang M, Hildebrand KA. The mast cell stabilizer ketotifen reduces joint capsule fibrosis in a rabbit model of post-traumatic joint contractures. Inflamm Res. 2012; 61: 285-292. doi: 10.1007/s00011-011-0409-3

77. Ehrlich HP. A snapshot of direct cell-cell communications in wound healing and scarring. Adv Wound Care (New Rochelle). 2013; 2: 113-121. doi: 10.1089/wound.2012.0414

78. Hei ZQ, Gan XL, Huang PJ, Wei J, Shen N, Gao WL. Influence of ketotifen, cromolyn sodium, and compound 48/80 on the survival rates after intestinal ischemia reperfusion injury in rats. BMC Gastroenterol. 2008; 8: 42. doi: 10.1186/1471-230X-8-42

79. Kalia N, Brown NJ, Wood RF, Pockley AG. Ketotifen abrogates local and systemic consequences of rat intestinal ischemia-reperfusion injury. J Gastroenterol Hepatol. 2005; 20: 1032-1038. doi: 10.1111/j.1440-1746.2005.03767.x

80. Sanchez-Patan F, Aller MA, Cuellar C, et al. Mast cell inhibition by ketotifen reduces splanchnic inflammatory response in a portal hypertension model in rats. Exp Toxicol Pathol. 2008; 60: 347-355. doi: 10.1016/j.etp.2008.03.008

81. Karmeli F, Eliakim R, Okon E, Rachmilewitz D. Gastric mucosal damage by ethanol is mediated by substance P and prevented by ketotifen, a mast cell stabilizer. Gastroenterology. 1991; 100: 1206-1216.

82. Eliakim R, Karmeli F, Rachmilewitz D. Ketotifen-Old drug, new indication: reduction of gastric mucosal injury. Scand J Gastroenterol. 1993; 28: 202-204. doi: 10.3109/00365529309096072

83. Ting S. Ketotifen and systemic mastocytosis. J Allergy Clin Immunol. 1990; 85: 818. doi: 10.1016/0091-6749(90)90205-i

84. Póvoa P, Ducla-Soares J, Fernandes A, Palma-Carlos AG. A case of systemic mastocytosis: therapeutic efficacy of ketotifen. J Intern Med. 1991; 229: 475-477. doi: 10.1111/j.1365-2796.1991.tb00379.x

85. Graves L, III, Stechschulte DJ, Morris DC, Lukert BP. Inhibition of mediator release in systemic mastocytosis is associated with reversal of bone changes. J Bone Miner Res. 1990; 5: 1113-1119. doi: 10.1002/jbmr.5650051104

86. Riccardi VM. Hiding in Plain Sight: A consideration of NF1-Associated Hypovitaminosis D and its treatment. J Genet Syndromes Gene Therapy. 2014. doi: 10.4172/2157-7412.1000223

87. Mena E, Brookstein JJ, Holt JF, Fry WJ. Neurofibromatosis and renovascular hypertension in children. AJR. 1973; 118: 39-45. doi: 10.2214/ajr.118.1.39

88. Geller M, Ribeiro MG, Araujo AP, de Oliveira LJ, Nunes FP. Serum IgE levels in neurofibromatosis 1. Int J Immunogenet. 2006; 33: 111-115. doi: 10.1111/j.1744-313X.2006.00579.x

89. Nakayama J, Sato C, Imafuku S. In vitro responses of neurofibroma fibroblasts, mast cells and Schwann cells obtained from patients with neurofibromatosis 1 to 308-nm excimer light and/or vitamin D. J Dermatol. 2013; 40. doi: 10.1111/1346-8138.12242

LATEST ARTICLES

Unraveling the Mysteries of Type-A Aortic Dissection Using POCUS/Echocardiography

Syeda Rukh*, Sathyanarayana Machani and Milind Awale

doi.

Blood Sample from the Patient

Hypertriglyceridemia-Induced Pancreatitis: A Case Report and Literature Review

Maarten Bulterys, Melvin Willems* and Agnes Meersman

doi.

From Neck Pain to a Life-Threatening Condition: A Case Report

Floris Vandewoude* and Sören Verstraete

doi.

LATEST ARTICLES