Effect of radiation on the sensori neural auditory system and the clinical implications 2

190 559 0
Effect of radiation on the sensori neural auditory system and the clinical implications 2

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

1. INTRODUCTION Radiation-induced radiation-induced sensori-neural hearing loss (SNHL) had long been recognized as a complication of radiotherapy (RT) for head and neck tumours, if the auditory pathways had been included in the radiation fields. It is believed that radiation therapy was a much larger etiologic factor of hearing loss than suspected and should clearly be recognized as a major factor in the etiology of adult hearing disorders (Mencher et al, 1995). The incidence of SNHL after radiotherapy for nasopharyngeal carcinoma (NPC) has been reported to be as high as 24% (Kwong et al, 1996). NPC is common among the Chinese and the main modality of treatment for NPC is RT. NPC is therefore common in Singapore, with a prevalence rate of 10.8 and 3.7 (agestandardized) per 100,000 per year for males and females respectively (Seow et al, 2004). In conventional RT of tumours located in the head and neck region, the auditory pathways are often included in the radiation fields. In NPC, many patients with post-RT hearing loss are encountered, both conductive and sensori-neural in nature (Low & Fong, 1998). While conductive hearing losses can normally be effectively overcome by wearing appropriate hearing aids, the use of hearing aids in SNHL may possibly result in sounds that are amplified but distorted (Low, 2005). Therefore, radiation-induced SNHL is of particular concern in Singapore, where the post-treatment quality of life is increasingly being emphasized. Today, we are faced with a number of clinical issues related to the effects of radiation on the sensori-neural audiory system. Do we know enough about these effects, so as to offer feasible solutions to the important clinical problems? Specifically: 1. Can the high incidence of radiation-induced SNHL be reduced after RTof head and neck tumours, if the auditory pathways had to be included in the radiation fields? This may be possible if the cellular and molecular basis of radiation-induced ototoxicity is known. 2. In patients with significant SNHL and whose auditory pathways had been irradiated before, cochlear implantation may be clinically indicated to restore hearing. In cochlear implantation, a pre-requisite for a successful outcome is intactness of the retro-cochlear pathways. In conventional modern-day RT, even with effective shielding of the brainstem, the cochlear nerve and ganglion are normally still at risk. Therefore, will radiation damage the retro-cochlear pathways, such that cochlear implants cannot work? 3. Combined chemo-radiotherapy using cisplatin (CDDP) has increasingly been used to treat advanced head and neck cancers like NPC (Wee J et al, 2005; Plowman, 2002). As CDDP is also known to be ototoxic, patients receiving combined radiation and CDDP therapy may be at high risk of losing significant hearing. Therefore, will the usefulness of combined therapy in head and neck cancers, be limited by unacceptable synergistic ototoxic effects? The relevant literature on the effects of radiation on the sensori-neural auditory system and related topics will be reviewed, focusing on the issues that have important clinical relevance. This thesis aims to address some of the gaps in knowledge that have the potential to improve clinical outcomes in the immediate and near future. 2. LITERATURE REVIEW 2.1 Characteristics of radiation-induced SNHL 2.1.1. Animal studies The effects of radiation on the inner ear was documented as early as 1905, when Ewald placed radium beads in the middle ear of pigeons and noted labyrinthine symptoms. Girden & Culler (1933) were first to evaluate the effects of radiation on hearing. Experimenting on dogs subjected to various X ray doses, a hearing impairment averaging 5.5 dB was recorded. Novotny (1951) studied the ionizing effects of radiation in guinea pigs and found a hearing impairment of about 8.4dB at 4000 Hz. Kozlov (1959) also noted a hearing imparment of 3.9-9.1dB over 500-8000 Hz in guinea pigs. Gamble et al (1968) subjected the ears of 50 guinea pigs to 500-6000 rads of radiation and found decreased sensitivity of cochlear microphonics of 20-40dB, particularly in the high frequencies. This was consistent with microscopic findings by Keleman (1963), who demonstrated damage to Organ of Corti and cochlear duct in rats, after doses of 100-3000 rads. Tokimoto & Kanagawa (1985) in their study on guinea pigs concluded that even in the absence of gross anatomical cochlear changes, post-irradiation functional hair cell defects could exist Histologically, Keleman (1963) studied temporal bones in rats which had received 1003000 rads of radiation. Haemorrhage was noted to be the most prominent finding with destruction of the cochlear duct, organ of corti and its surrounding elements. Gamble et al (1968) reported early changes of the stria vascularis, accompanied by significant inflammatory responses in the inner ears of guinea pigs which had received 6000 rads of radiation. 2.1.2 Human studies Several clinical studies have recorded SNHL in patients who have had RT for head and neck malignancies, where inner ear structures were included in the radiation fields. Leach (1965) observed SNHL in some of the 56 patients who had received 3000-12000 rads of RT for different head and neck cancers. Morretti (1976) retrospectively studied 137 postirradiated NPC patients, and found to have SNHL of at least 10dB. However, there had also been studies which suggested that radiation did not result in significant SNHL (Evans et al, 1988). In a systematic review of the literature, Raajmakers & Engelen (2002) explained that the conflicting results were attributed to variations in patient groups, size, study design, follow-up period, radiotherapy techniques and presentation of audiometric results. From the pooled data generated from their systematic review, they concluded that about one-third of patients who had received 70 Gy in Gy per fraction near the inner ear, developed hearing loss of 10dB or more at kHz. Schnecht & Karmody (1966) reported the histological features of a deafened man who had received 5,220 rads of radiation to the region of the ears several years ago. Degeneration of the Organ of Corti was noted, with atrophy of the basilar membrane, spiral ligament and stria vascularis. Progressive hearing loss across the various frequencies had been attributed to obliterating endarteritis and eventual fibrosis, leading to vascular compromise (Morreti, 1976). 2.1.3 Prevalence & epidemiology Mencher et al (1995) lamented that it was difficult to establish the exact prevalence of radiation-induced hearing loss because statistics relating to ototoxicity was often not well kept or easily interpreted. Not only was hearing not often considered important in the face of life-threatening diseases, hearing losses were frequently not recorded in those who did not survive. Clinical studies had reported varying prevalence, depending on factors such as dose, period of follow-up and definition/criteria used for hearing loss (Raajmakers & Engelen, 2002). In NPC where the radiation dose received by the ear is relatively high, the incidence of post-RT SNHL had been reported to be as high as 24% (Kwong et al, 1996). In SNHL after RT in NPC patients, sex and age were found to be independent prognostic factors (Kwong et al, 1996). Males were noted to be more susceptible than females in developing SNHL after radiation. Older patients were at greater risk, as pre-existing degenerative changes could have made them more vulnerable to radiation injury (Wang et al, 2003). Post-RT middle ear effusion was identified to be another predicting factor, as toxic materials from chronic inflammation could affect the inner ear (Oh et al, 2004). However, it is argued that the development of post-RT middle ear effusion could have been just another manifestation of radiation damage related to individual variation in susceptibility to radiation (Kwong et al, 1996). 2.1.4 Effect of radiation dose Gamble et al (1968) found in guinea pigs that the extent of inner ear injury correlated with the radiation dose applied. Bohne et al (1985) confirmed in chinchillas that the higher the radiation dose received, the greater the damage to the inner ear. A systematic review on human studies reported increasing loss with increasing dose, starting at about 40 Gy applied in Gy per fraction (Raajmakers & Engelen, 2002). In fact, guidelines for tolerance doses in normal tissues are being used in clinical practice (Emami et al, 1991). It is noteworthy that early human studies had reported the effects of very high doses of radiation to the ears, doses that are no longer used in clinical practice today (Thibadoux et al, 1980; Talmi et al, 1988). Although these could have potentially provided rare opportunities to study the effect of excessively high doses of radiation on the inner ear in humans, documentation of the audiometric data was so poor that it was impossible to draw conclusions on the relationship between high radiation dose and SNHL. 2.1.5 High frequency hearing loss Hearing in the high frequencies were consistently found to be more affected than hearing in the lower frequencies after irradiation (Raajmakers & Engelman, 2002; Talmi et al, 1989). This corresponded to histological observations made in animals that the basal part of the cochlea (which respond to higher frequency sounds) was usually more damaged by radiation than the apical part. Keleman (1963) demonstrated in rats that the apical turn of the cochlea was least affected by radiation doses of 100-3000 rads. Winter (1970) similarly reported in guinea pigs that post-irradiated apical hair cells remained intact while the outer hair cells of the basal turns were affected. Early threshold shifts at high stimulus frequencies are indicators of probable subsequent shifts to low frequencies (Schell et al, 1989). 2.1.6 Early vs late onset Traditionally, radiation-induced SNHL is regarded to have either early or late onsets (Talmi et al, 1989). The existence of early-onset radiation-induced cochlear damage had been convincingly demonstrated in both animals and humans. Winter (1970) reported in guinea pigs, hair cell damage as early as hrs following radiation of 4,000-7,000 rads. Tokimoto & Kanagawa (1985) demonstrated in guinea pigs that sensori-neural loss appeared 3-10 hrs depending on dose administered, and outer hair cells in the basal turn of the cochlea were destroyed about hrs after radiation. In humans, SNHL often occurred near the end or shortly after the completion of fractionated RT (Linskey & Johnstone, 2003). Early-onset SNHL due to inflammatory causes or transient functional disturbances in the stria vascularis, may recover with time (Kwong et al, 1996; Linskey & Johnstone, 2003). In late-onset hearing loss, Schuknecht & Karmordy (1966) observed marked atrophy of the cochlear stria vascularis in a patient who had developed hearing loss years after radiotherapy (5200 rads) for carcinoma of the ear. Grau et al (1991) studied 22 NPC patients with well-documented pre- and post-RT hearing levels over 7-84 months, and found SNHL (especially the higher frequencies) developing 12 months post-RT. Merchant et al (2004) were of the opinion that radiation-induced SNHL could only occur years after irradiation. Delayed-onset hearing loss was found to correlate with age, preexisting SNHL and radiation dosage (Honore et al, 2002). Late-onset radiation-induced SNHL had generally been attributed to progressive vascular compromise from radiationinduced vasculitis obliterans (Morretti, 1976). However, after a review of the relevant literature, Sataloff et al (1994) were not convinced that late-onset hearing loss existed at all. They argued that although significant hearing losses over the whole spectrum of the speech range were often seen several years after radiation, there was little or no convincing evidence to support the notion that hearing loss developing several years following radiation was causally related to radiation therapy itself. In summary, although early-onset radiation-induced SNHL and its progressive nature have generally been well accepted, late-onset radiation-induced SNHL attributed to progressive vascular compromise has not been convincingly shown. Therefore, late-onset radiation-induced hearing loss may well be a later manifestation of progressive earlyonset hearing loss. 2.2 Effect of radiation on retro-cochlear pathways 2.2.1 The sensori-neural auditory pathways According to Hackney (1987), the hair cells of the Organ of Corti transduce vibrations within the cochlea into neural signals. Outer hair cells are contractile and may contribute to mechanical feedback processes, whilst the inner hair cells are apparently the primary sensory cells being innervated by the majority of the afferent fibres. These run in the cochlear nerve to the brain stem where they bifurcate, projecting cochleotopically to the dorsal and ventral cochlear nuclei. A divergence continued in the main routes taken by the ascending pathways; one runs bilaterally from the ventral cochlear nucleus to the superior olivary complex and then to the inferior colliculi, the other runs from the dorsal cochlear nucleus to the contralateral inferior colliculus. Fibres from the brainstem nuclei travelling to each inferior colliculus form a tract, the lateral lemniscus, and may make contact with one of the nuclei within it. The pathway continues to the medial geniculate bodies and on to the auditory cortex, preserving its cochleotopicity at all levels. A descending system parallels the ascending system throughout. The presence of commissural and decussating connections from the level of the brainstem onwards, provides the anatomical basis for the analysis of binaural information. The division of the pathway forms the anatomical substrate for the parallel processing of different features of the auditory environment. 10 Mencher GT, Novotny G, Mencher L, Gulliver M (1995). Ototoxicity and irradiation: additional etiologies of hearing loss in adults. J Am Acad Audiol; 6:351-357. Merchant TE, Gould CJ, Xiong X, et al (2004). Early neuro-otologic effects of threedimensional irradiation in children with primary brain tumors. Int J Radiat Oncol Biol Phys 15:1194-207. Miettinen S, Laurikainen E, Johansson R, et al (1997). Radiotherapy enhanced ototoxicity of cisplatin in children. Acta Otolaryngol (Stockh) Suppl 529: 90-94. Miller FD, Pozniak CD, Walsh GS (2000). Neuronal life and death: an essential role for the p53 family.Cell Death Differ;7:880-8. Miller LJ, Marx J (1998). Apoptosis. Science; 281:1301. Momiyama J, Hashimoto T, Matsubara A, Futai K, Namba A, Shinkawa H (2006). Leupeptin, a calpain inhibitor, protects inner ear hair cells from aminoglycoside ototoxicity. Tohoku J Exp Med;209:89-97 Moretti JA (1976). Sensori-neural hearing loss following radiotherapy to the nasopharynx. Laryngoscope; 86:598-602. 175 Morris EJ, Keramaris E, Rideout HJ, Slack RS, Dyson NJ, Stefanis L, Park DS (2001). Cyclin-dependent kinases and P53 pathways are activated independently and mediate Bax activation in neurons after DNA damage. J Neurosci;21:5017-26. Morrison RS, Kinoshita Y, Johnson MD, Guo W, Garden GA (2003). p53-dependent cell death signaling in neurons.Neurochem Res;28:15-27. Naderi S, Hunton IC, Wang JY (2002). Radiation dose-dependent maintenance of G(2) arrest requires retinoblastoma protein. Cell Cycle;1:193-200. Nagata S. Apoptosis. http://claim.springer.de/EncRef/CancerResearch/samples/0003.htm. Nakajima T (2006). Signaling cascades in radiation-induced apoptosis: Roles of protein kinase C in the apoptosis regulation.Med Sci Monit;12:RA220-224. Narang H, Krishna M (2004). .Mitogen-activated protein kinases: specificity of response to dose of ionizing radiation in liver. J Radiat Res (Tokyo);45:213-20. Nightingale S, Schofield IS, Dawes PJ (1984). Visual, cortical somatosensory and brainstem auditory evoked potentials following incidental irradiation of the rhombencephalon. J Neurol Neurosurg Psychiatry;47:91-3 176 Nomura R, Hattori T, Yanagita N (1997). Radiation tolerance of the cochlear nerve at the gamma-knife in rabbits.Auris Nasus Larynx;24:341-9. Novotny O (1951). Sull’azione dei raggi x sulla chiocciola della cavia. Arch Ital Otol Rhinol Lar; 62:15-9. Oesterle EC, Cunningham DE, Westrum LE, Rubel EW (2003). Ultrastructural analysis of [3H]thymidine-labeled cells in the rat utricular macula J Comp Neurol;463:177-95. Ogawa Y, Kobayashi T, Nishioka A, Kariya S, Hamasato S, Seguchi H, Yoshida S (2003). Radiation-induced reactive oxygen species formation prior to oxidative DNA damage in human peripheral T cells. Int J Mol Med;11:149-52. Oh YT, Kim CH, Choi JH, Kang JH, Chun M (2004). Sensory neural hearing loss after concurrent cisplatin and radiation therapy for nasopharyngeal carcinoma. Radiother Oncol; 72:79-82. Ohlemiller KK, McFadden SL, Ding DL, Lear PM, Ho YS (2000). Targeted mutation of the gene for cellular glutathione peroxidase (Gpx1) increases noise-induced hearing loss in mice. Assoc Res Otolaryngol;1:243-54. O'Leary SJ, Klis SF (2002). Recovery of hearing following cisplatin ototoxicity in the guinea pig. Anticancer Res;22:1525-8. 177 Ondrey FG, Greig JR, Herscher L (2000). Radiation dose to otologic structures during head and neck cancer radiation therapy. Laryngoscope; 110:217-21. Pagoria D, Geurtsen W (2005) The effect of N-acetyl-l-cysteine and ascorbic acid on visible light-irradiated camphorquinone /N,N-dimethyl-p-toluidine-induced oxidative stress in two immortalized cell lines. Biomaterials; 26:6136-42 Parker MA, Cotanche DA (2004). The potential use of stem cells for cochlear repair. Audiol Neurootol;9:72-80. Pena LA, Fuks Z, Kolesnick R (1997). Stress-induced apoptosis and the sphingomyelin pathway.Biochem Pharmacol;53:615-21. Pirvola U, Xing-Qun L, Virkkala J, Saarma M, Murakata C, Camoratto AM, Walton KM, Ylikoski J (2000). Rescue of hearing, auditory hair cells, and neurons by CEP1347/KT7515, an inhibitor of c-Jun N-terminal kinase activation.J Neurosci;20:43-50. Plowman PN (2002). Post-radiation sensori-neural hearing loss. Int J Rad Oncol Biol Phys; 589-91. Potapova O, Basu S, Mercola D, Holbrook NJ (2001). Protective role for c-Jun in the cellular response to DNA damage. J Biol Chem;276:28546-53. 178 Previati M, Lanzoni I, Corbacella E, Magosso S, Giuffre S, Francioso F, Arcelli D, Volinia S, Barbieri A, Hatzopoulos S, Capitani S, Martini (2004). RNA expression induced by cisplatin in an organ of Corti-derived immortalized cell line. Hear Res;196:818. Przyborski SA, Morton IE, Wood A, Andrews PW (2000). Developmental regulation of neurogenesis in the pluripotent human embryonal carcinoma cell line NTERA-2. Eur J Neurosci;12:3521-8. Raajmakers E, Engelen AM (2002). Is sensorineural hearing loss a possible side effect of nasopharyngeal and parotid irradiation? A systematic review of the literature. Radiotherapy Oncol; 65: 1-7. Rafael RC, Jose Ramon GB, Julia B, Alejandro MM, Almudena T (2004). Supporting cells as a target of Cisplatinum-induced inner ear damage: therapeutic implications. Laryngoscope; 114:533-7. Raff M (2003). Adult stem cell plasticity: fact or artifact? Annu Rev Cell Dev Biol;19:122. Raff M (1998). Cell suicide for beginners. Nature; 396: 119-122. 179 Ravi R, Somani SM, Rybak LP (1995). Mechanism of cisplatin ototoxicity: antioxidant system. Pharmacol Toxicol.;76:386-94 Richardson GP, Russell IJ (1991). Cochlear cultures as a model system for studying aminoglycoside induced ototoxicity. Hear Res;53:293-311 Rivolta MN, Holley MC (2002). Cell lines in inner ear research. J Neurobiol;53:306-18. Rubel EW, Dew LA, Roberson DW (1995). Mammalian vestibular hair cell regeneration. Science;267:701-7. Ruben RJ (1967). Development of the inner ear of the mouse: a radioautographic study of terminal mitoses. Acta Otolaryngol;Suppl 220:1-44. Ryals BM, Rubel EW (1988). Hair cell regeneration after acoustic trauma in adult Coturnix quail. Science;240:1774-6. Ryan A (2006). Challenges for gene therapy of the inner ear. Bionics and regeneration of the ear. 7th International Academic Conference of Immunobiology in Otorhinolaryngologylogy Abstract Book, Melbourne, pp 17 Rybak LP, Whitworth CA (2005). Ototoxicity: therapeutic opportunities. Drug Discov Today;10:1313-21. 180 Sataloff RT, Rosen DC (1994). Effects of cranial irradiation on hearing acuity: a review of the literature. Am J Otol;15:772-80. Scarpidis U, Madnani D, Shoemaker C, Fletcher CH, Kojima K, Eshraghi AA, Staecker H, Lefebvre P, Malgrange B, Balkany TJ, Van De Water TR (2003). Arrest of apoptosis in auditory neurons: implications for sensorineural preservation in cochlear implantation. Otol Neurotol;24:409-17. Schacht J, Sha SH, Qiu JH (2006). Gentamicin attenuates aminoglycoside ototoxicity: from laboratory to clinic. Bionics and regeneration of the ear. 7th International Academic Conference of Immunobiology in Otorhinolaryngologylogy Abstract Book, Melbourne, pp 7. Schell M, McHaney VA, Green AA, et al (1989). Hearing loss in children and young adults receiving cisplatin with and without prior cranial irradiation. J Clin Oncol; 7:754760. Schreck R, Rieber P, Baeuerle PA (1991). Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-kappa B transcription factor and HIV-1. EMBO J;10:2247-58. 181 Schuknecht HF, Karmody CS (1966). Radionecrosis of the temporal bone. Laryngoscope; 76: 1416-28. Seidman MD, Vivek P (2004). Intratympanic treatment of hearing loss with novel and traditional agents. Otolaryngol Clin North Am;37:973-90. Seigel GM (1999). The golden age of retinal cell culture. Mol Vis;5:4. Senften M, Schwander M, Kazmierczak P, Lillo C, Shin JB, Hasson T, Geleoc GS, Gillespie PG, Williams D, Holt JR, Muller U (2006). Physical and functional interaction between protocadherin 15 and myosin VIIa in mechanosensory hair cells. J Neurosci; 26:2060-71. Sentman CL, Shutter JR, Hockenbery D, Kanagawa O, Korsmeyer SJ (1991). bcl-2 inhibits multiple forms of apoptosis but not negative selection in thymocytes. Cell;67:879-88. Seow A, Koh WP, Chia KS, Shi LM, Lee HP, Shanmugaratnam K (2004). Nasopharynx: In trends in cancer incidence in Singapore 1968-2002. Report No 6, Singapore Cancer Registry, Singapore pp 88-9. 182 Sha SH, Schacht J (2000). Antioxidants attenuate gentamicin-induced free radical formation in vitro and ototoxicity in vivo: D-methionine is a potential protectant. Hear Res;142:34-40. Sha SH, Taylor R, Forge A, Schacht J (2001). Differential vulnerability of basal and apical hair cells is based on intrinsic susceptibility to free radicals. Hear Res;155:1-8. Shinomiya N (2001). New concepts in radiation-induced apoptosis: 'premitotic apoptosis' and 'postmitotic apoptosis'. J Cell Mol Med;5:240-53. Shou J, Zheng JL, Gao WQ (2003). Robust generation of new hair cells in the mature mammalian inner ear by adenoviral expression of Hath 1. Mol Cell Neurosci;23:169-79. Skinner R, Pearson ADJ, Amineddine HA, et al (1990). Ototoxicity of cisplatinum in children and adolescent. Br J Cancer; 61:927-931. Smolders JW (1999). Functional recovery in the avian ear after hair cell regeneration. Audiol Neurotol;4:286-302. Stefanis L (2005). Caspase-dependent and -independent neuronal death: two distinct pathways to neuronal injury. Neuroscientist;11:50-62. 183 Strasser A, Harris AW, Jacks T, Cory S (1994). DNA damage can induce apoptosis in proliferating lymphoid cells via p53-independent mechanisms inhibitable by Bcl-2. Cell;79:329-39. Strom E, Sathe S, Komarov PG, Chernova OB, Pavlovska I, Shyshynova I, Bosykh DA, Burdelya LG, Macklis RM, Skaliter R, Komarova EA, Gudkov AV (2006). Smallmolecule inhibitor of p53 binding to mitochondria protects mice from gamma radiation. Nat Chem Biol;2:474-9. Sweetow RW, Will TI (1993). Progression of hearing loss following the completion of chemotherapy and radiation therapy: case report. J Am Acad Audiol; 4:360-363. Takahashi A, Matsumoto H, Yuki K, Yasumoto J, Kajiwara A, Aoki M, Furusawa Y, Ohnishi K, Ohnishi T (2004). High-LET radiation enhanced apoptosis but not necrosis regardless of p53 status. Int J Radiat Oncol Biol Phys;60:591-7 Talmi Y, Kalmanovitch M, Zohar Y (1988). Thyroid carcinoma, cataract, and hearing loss in a patient afer irradiation for facial hemangioma. J Laryngol Otol; 102: 91-2. Talmi YP, Finkelstein Y, Zohar Y (1989). Postirradiation hearing loss. Audiology;28:121-6. 184 Tan DTH, Liu YP, Sun L (2000). Flow cytometer measurements of DNA content in primary and recurrent ptergia. Inv Ophothol & Vis Sci;41:1684-6. Tateya I, Nakagawa T, Iguchi F, Kim TS, Endo T, Yamada S, Kageyama R, Naito Y, Ito J (2003). Fate of neural stem cells grafted into injured inner ears of mice.Neuroreport;14:1677-81. Thibadoux GM, Pereira WV, Hodges JM, Aur RJ (1980). Effects of cranial radiation on hearing in children with acute lymphocytic leukemia. J Pediatr;96:403-6. Thorne PR (2006). Oxidative stress- its role in cochlear injury. Bionics and regeneration of the ear. 7th International Academic Conference of Immunobiology in Otorhinolaryngologylogy Abstract Book, Melbourne, pp Tokimoto T, Kanagawa K (1985). Effects of x-ray irradiation on hearing in guinea pigs. Acta Otolaryngol (Stockh); 100:266-72. Truman JP, Gueven N, Lavin M, Leibel S, Kolesnick R, Fuks Z, Haimovitz-Friedman A (2005). Down-regulation of ATM protein sensitizes human prostate cancer cells to radiation-induced apoptosis.J Biol Chem;280:23262-72. Usami S (1996). Differential cellular distribution of glutathione – an endogenous antioxidant in the guinea pig inner ear. Brain Res; 743:337-40. 185 Valavanis C, Hu Y, Yang Y, Osborne BA, Chouaib S, Greene L, Ashwell JD, Schwartz LM (2001). .Model cell lines for the study of apoptosis in vitro. Methods Cell Biol;66:417-36. Van De Water TR, Lallemend F, Eshraghi AA, Ahsan S, He J, Guzman J, Polak M, Malgrange B, Lefebvre PP, Staecker H, Balkany TJ (2004). Caspases, the enemy within, and their role in oxidative stress-induced apoptosis of inner ear sensory cells. Otol Neurotol;25:627-32. Van der Hulst RJAM, Dreschlear WA, Urbanus NAM (1988). High frequency audiometry in prospective clinical research of ototoxicity due to platinum derivatives. Ann Otol Rhinol Laryngol; 97:133-7. Van Eck AT, Horstmann GA (2005). Increased preservation of functional hearing after gamma knife surgery for vestibular schwannoma. .J Neurosurg;102 Suppl:204-6. Verheij M, Bartelink H (2000). Radiation-induced apoptosis. Cell Tissue Res;301:13342. Verheij M, Bose R, Lin XH, Yao B, Jarvis WD, Grant S, Birrer MJ, Szabo E, Zon LI, Kyriakis JM, Haimovitz-Friedman A, Fuks Z, Kolesnick RN (1996). Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis. Nature;380:75-9. 186 Walker DA, Pillow J, Waters KD, et al (1989): Enhanced Cis-platinum ototoxicity in children with brain tumours who have received simultaneous or prior cranial irradiation. Med Pedia Oncol; 17:48-52. Wang J, Ladrech S, Pujol R, Brabet P, Van De Water TR, Puel JL (2004). Caspase inhibitors, but not c-Jun NH2-terminal kinase inhibitor treatment, prevent cisplatininduced hearing loss. Cancer Res;64:9217-24. Wang J, Van De Water TR, Bonny C, de Ribaupierre F, Puel JL, Zine A (2003). A peptide inhibitor of c-Jun N-terminal kinase protects against both aminoglycoside and acoustic trauma-induced auditory hair cell death and hearing loss. J Neurosci;23:8596607. Wang LF, Kuo WR, Ho KY, et al (2003). Hearing loss in patients with nasopharyngeal carcinoma after chemotherapy and radiation. Kaoshsiung J Med Sci; 19:163-169. Ward WD (1973). Noise-induced hearing damage. In Paparella MM, Shomrick DA (eds): Otolaryngology, Vol 2. Philadelphia, Saunders, pp 337-390. Weatherly RA, Owens JJ, Catlin FI, et al (1991). Cis-platinum ototoxicity in children. Laryngoscope; 101:917-24. 187 Wee J, Tan EH, Tai BC, Wong HB, Leong SS, Tan T, Chua ET, Yang E, Lee KM, Fong KW, Tan HS, Lee KS, Loong S, Sethi V, Chua EJ, Machin D (2005). Randomized trial of radiotherapy versus concurrent chemoradiotherapy followed by adjuvant chemotherapy in patients with American Joint Committee on Cancer/International Union against cancer stage III and IV nasopharyngeal cancer of the endemic variety.J Clin Oncol;23:6730-8. Wee JTS, KhooTan HS, Chua EJ (1997). Radiotherapy. In: Nasopharyngeal Cancer (eds. Chong VH, Tsao SY), Armour Publishing, Singapore, pp 90-102. Winther FO (1970). X-ray irradiation of the inner ear of the guinea pig – an electron microscopic study of degenerating outer hair cells of the Organ of Corti. Acta Otolaryngol; 69:61-6. Wichmann A, Jaklevic B, Su TT (2006). Ionizing radiation induces caspase-dependent T but Chk2- and p53-independent cell death in Drosophila melanogaster. Proc Natl Acad Sci U S A;103:9952-7 Withers HR (1992). Biologic basis of radiation therapy. In: Principles and Practice of radiation oncology. 2nd edition. Peretz CA, Brady LW (eds.). JB Lippincott Co, Philadelphia, pp64-96. 188 Wright CG, Schaefer SD (1982). Inner ear histopathology in patients treated with cisplatinum. Laryngoscope;92:1408-13. Wu WJ, Sha SH, Schacht J (2002). Recent advances in understanding aminoglycoside ototoxicity and its prevention. Audiol Neurootol;7:171-4 Xiang H, Kinoshita Y, Knudson CM, Korsmeyer SJ, Schwartzkroin PA, Morrison RS (1998). Bax involvement in p53-mediated neuronal cell death. J Neurosci;18:1363-73. Yamada T, Ohyama H (1988). Radiation-induced interphase death of rat thymocytes is internally programmed (apoptosis).Int J Radiat Biol Relat Stud Phys Chem Med;53:6575. Ylikoski J, Xing-Qun L, Virkkala J, Pirvola U (2002). Blockade of c-Jun N-terminal kinase pathway attenuates gentamicin-induced cochlear and vestibular hair cell death.Hear Res;163:71-81. Zafarullah M, Li WQ, Sylvester J, Ahmad M (2003). Molecular mechanisms of Nacetylcysteine actions. Cell Mol Life Sci;60:6-20. Zhang M, Liu W, Ding D, Salvi R (2003). Pifithrin-alpha suppresses p53 and protects cochlear and vestibular hair cells from cisplatin-induced apoptosis. Neuroscience;120:191-205. 189 Zheng J, Shen W, He DZ, Long KB, Madison LD, Dallos P (2000). Prestin is the motor protein of cochlear outer hair cells. Nature;405:149-55. Zheng JL, Frantz G, Lewis AK, Sliwkowski M, Gao WQ (1999). Heregulin enhances regenerative proliferation in postnatal rat utricular sensory epithelium after ototoxic damage.J Neurocytol;28:901-12. Zheng JL, Gao WQ (2000). Overexpression of Math1 induces robust production of extra hair cells in postnatal rat inner ears.Nat Neurosci;3:580-6. Zine A,van de Water TR (2004). The MAPK/JNK signalling pathway offers potential therapeutic targets for the prevention of acquired deafness.Curr Drug Targets CNS Neurol Disord;3:325-32. 190 [...]... demonstrated in rabbits, loss of ganglion cell bodies and cochlear ganglia after more than 40 Gy of gamma radiation On the other hand, Keleman (1963) did not observe any effect on the cochlear nerve fibres after single doses of photon radiation less than 20 Gy As the effects of radiation on nervous tissues could be related to dose, Bohne et al (1985) studied the effects of ionising radiation on the. .. seen that the existing literature gave conflicting views on the effect of radiotherapy on the retro-cochlear pathways It is pointed out that many of the studies were retrospective and were based on small numbers of patients Prospective studies on the long-term effects beyond 1 year after irradiation, have not been done 2. 3 Combined effects of radiation and CDDP on SNHL 2. 3.1 Combined chemo-radiotherapy... Although the brainstem is well shielded during RT, the retro-cochlear auditory pathways at the level of the spiral ganglia (located in the modiolus of the cochlea) and cochlear nerve remain at risk, as these structures are not effectively shielded during treatment There have been only limited studies on the effects of radiation on the spiral ganglia and auditory nerve, and the results have not been conclusive... 5,000 rads In these patients, 54% had pronounced deterioration of thresholds and/ or speech discrimination; BERA was abnormal in all but one ear However, because the post-irradiation hearing results are often confounded by 16 the tumour effect on the cochlear nerve (Kaplan et al, 20 03), acoustic neuroma is not a suitable clinical model to study the effects of radiation on the retro-cochlear auditory pathways... (Adrian, 20 02) The Bcl -2 family consists of a group of proteins that function as a checkpoint for cell death and survival signals at the level of the mitochondria Bcl -2 family members can be characterized as either anti-apoptotic (eg Bcl -2) or pro-apoptotic (eg Bax) Upon receiving the stress signal, the proapoptotic proteins in the cytoplasm, BAX and BID, bind to the outer membrane of the mitochondria... based on their prospective study of 22 patients evaluated prior to and 7-84 months after radiotherapy for NPC They found that auditory brainstem evoked responses in 4 patients were severely abnormal and 2 had clinical signs of brainstem dysfunction Grau et al (19 92) had shown by dose response analysis, a correlation between total radiation doses received by the brainstem and the incidence of pathologic... 20 05) 32 2.5 Radiation- induced apoptosis 2. 5.1 Radiobiology The biological effects of radiation can be mediated via a direct or an indirect mechanism (Withers, 19 92) When radiation is absorbed in a biological material, the atoms in the cellular target itself may be ionized or excited, thus initiating the chain of events that leads to a biological change This is the so-called direct action of radiation; ... chincillas to 40 to 90 Gy of radiation, fractionated at 2 Gy per day The animals were sacrificed two years after completion of treatment and the temporal bones were studied microscopically Dose dependent degeneration of sensory and supporting cells as well as loss of 8th nerve fibres in the Organ of Corti, were observed In ears exposed to 40-50Gy of radiation, the incidence of nerve fibre damage was... Vougiouka, 20 03) The hydrolysis product is believed to be the active species reacting mainly with glutathione in the cytoplasm and the DNA in the nucleus, thus inhibiting replication, transcription and other nuclear functions A number of additional properties of CDDP are now emerging, including activation of signal transduction pathways leading to apoptosis Firing of such pathways 18 may originate at the. .. chemo-radiotherapy is increasingly being used clinically to treat advanced head and neck cancers In RT of tumours in the head and neck region, the auditory pathways are often included in the radiation fields and radiation- induced SNHL may result CDDP, widely used as an effective anti-neoplastic drug for these cancers, is also well known to cause ototoxicity Therefore, in combined therapy, the synergistic . Gy of gamma radiation. On the other hand, Keleman (1963) did not observe any effect on the cochlear nerve fibres after single doses of photon radiation less than 20 Gy. As the effects of radiation. and were based on small numbers of patients. Prospective studies on the long-term effects beyond 1 year after irradiation, have not been done. 2. 3 Combined effects of radiation and CDDP on. number of clinical issues related to the effects of radiation on the sensori-neural audiory system. Do we know enough about these effects, so as to offer feasible solutions to the important clinical

Ngày đăng: 14/09/2015, 10:44

Từ khóa liên quan

Mục lục

  • 1. INTRODUCTION

    • 2.7 Using cochlear cell-lines to study ototoxicity

    • 3. OBJECTIVES

    • 4. TO STUDY THE EFFECT OF RADIATION ON RETRO-COCHLEAR PATHWAYS

      • 4.1 Abstract

          • 4.3.1 Patients

            • 4.3.2 Radiotherapy Technique

              • 4.3.3 Radiation Dose Measurements

              • 4.4.1 Audiological Effects of Radiation

              • 4.4.2 Radiation Doses to the Cochlea and Auditory Nerve

                • 5.1 Abstract

                    • 5.3.1 Patients

                    • Concurrent Chemo-radiotherapy

                    • THERAPY DOSE ROUTE DAYS

                      • Radiotherapy 200 cGy / day 35 daily fractions

                        • Adjuvant Chemotherapy

                        • THERAPY DOSE ROUTE DAYS

                        • 5.3.6 Statistical methods

                        • 5.4 Results

                          • Table 5. Characteristics of patients in each treatment group

                            • Sex

                              • Race

                                • 1 Week

                                • 6 Months

                                • 1 Year

                                  • 2 Years

                                    • Beyond the follow-up period of this study, the post-treatment sensori-neural hearing in both groups is expected to further decline over time, with the onset of delayed radiation-induced hearing loss (Merchant et al, 2004; Ho et al, 1999; Sweetow & Will, 1993). Both RT & CDDP are also reported to be progressive in nature (Wang et al, 2003; Sweetow & Will, 1993). It remains to be seen how the post-treatment sensori-neural hearing outcomes between the 2 groups will differ in the longer term, as the rate and degree of deterioration may not necessarily be the same. Nevertheless, it would not be unreasonable to expect over the longer term, the sensori-neural hearing of patients in the chemo-radiotherapy group to remain significantly poorer than that of patients in the RT group; possibly to an even greater extent.

                                    • In conclusion, this study demonstrated that patients with nasopharyngeal carcinoma who had received combined RT and concurrent/adjuvant chemotherapy using CDDP experienced greater SNHL compared to those treated by RT alone, especially to high frequency sounds in the speech range. Combined therapy appeared to have allowed the plateau representing ototoxic destruction of outer hair cells to be reached at much lower levels of CDDP, than with chemotherapy alone. Further research should focus on the cellular and molecular processes of radiation-induced cell death in the cochlea.

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan