Accepted Manuscript Title: PHOTODYNAMIC THERAPY IN THE TREATMENT OF BRAIN TUMOURS. A FEASIBILITY STUDY Author: Vicente Vanaclocha Manuel Sureda Ignacio Azinovic Joseba Rebollo Rosa Ca˜no´ n Nieves Saiz Sapena Francisco Garc´ıa Cases Antonio Brugarolas PII: DOI: Reference:

S1572-1000(15)00069-1 http://dx.doi.org/doi:10.1016/j.pdpdt.2015.05.007 PDPDT 656

To appear in:

Photodiagnosis and Photodynamic Therapy

Received date: Revised date: Accepted date:

29-12-2014 7-5-2015 18-5-2015

Please cite this article as: Vanaclocha V, Sureda M, Azinovic I, Rebollo J, Ca˜no´ n R, Sapena NS, Cases FG, Brugarolas A, PHOTODYNAMIC THERAPY IN THE TREATMENT OF BRAIN TUMOURS. A FEASIBILITY STUDY, Photodiagnosis and Photodynamic Therapy (2015), http://dx.doi.org/10.1016/j.pdpdt.2015.05.007 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1 2

PDT is a local treatment modality without long-term systemic effects.

3 4

Chemo and RT continue to present important limitations in brain tumors

6

ip t

5

41 patients with a median age of 49, with primary brain tumors were treated with PDT

8

cr

7

Results are not inferior to those in literature, in spite of the characteristics of the patients

10

us

9

PDT can be considered as an adjunctive to other modalities in brain tumors

Ac ce p

te

d

M

an

11

1 Page 1 of 17

PHOTODYNAMIC THERAPY IN THE TREATMENT OF BRAIN TUMOURS. A

12

FEASIBILITY STUDY.

13

Vicente Vanaclocha1, Manuel Sureda2, Ignacio Azinovic2, Joseba Rebollo2, Rosa Cañón2,

14

Nieves Saiz Sapena1, Francisco García Cases2, Antonio Brugarolas2

15

1

16

Torrevieja, Torrevieja, Alicante, Spain

ip t

11

cr

Hospital 9 de Octubre, Valencia, Spain 2Plataforma de Oncología-Hospital Quirón

Corresponding Author: Manuel Sureda, MD, PhD

19

Plataforma de Oncología-Hospital Quirón Torrevieja,

20

c/Partida de la Loma s/n

21

03184 Torrevieja, Alicante, Spain

22

Tel +34651848837

23

[email protected]

M te

Background

Photodynamic therapy (PDT) constitutes a treatment modality that combines a photosensitizing agent with exposure to laser light in order to elicit phototoxic reactions that selectively destroy tumor cells and spare normal cells. PDT is a local treatment modality without long-term systemic effects. Its application can be repeated more than once to the same area without accumulative effects.

Ac ce p

25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46

d

24

an

18

us

17

Methods

Patients diagnosed with primary brain tumors were treated with PDT. Treatment consisted in administration of the photosensitizer followed by craniotomy, surgical resection and laser illumination of the surgical bed. Primary brain tumors received also temozolomide-based chemotherapy and radiotherapy (RT). Results

From May 2000 to December 2010, 41 patients (27 male, 14 female) with a median age of 49 years (range 13 to 70) diagnosed of primary brain tumors were included in the study. In 7 patients PDT was repeated at the time of the relapse. In 22 episodes PDT was part of the initial treatment of primary brain tumors and in 26 episodes was part of the treatment at relapse.

Median PFS observed was 10 months for GBM (95% confidence interval 5.7-14.3), 26 months for AA (95% CI 4.5-47.5), and 43 months for OD (95% CI 4.5-47.5). Median OS was 9 months for GBM (95% CI 2.3-15.7), 20 months for AA (95% CI 0.0-59) and 50 months for OD (95% CI 32.5-67.5). The apparent discrepancy between PFS and OS data is due to patients not censored for PFS because they die from causes other than tumor progression. Median OS since first diagnosis was 17 months for GBM (95% CI 15.2-17.8), 66 months for AA (95% CI 2.9-129.1) and 122 months for OD (95% CI 116.1-127.8). Side effects were mild and manageable. Conclusions

2 Page 2 of 17

47 48 49 50

This study confirms that PDT can be considered as an adjunctive to surgery and/or RT and chemotherapy in the treatment of brain tumors, excluding those patients with thalamic or brain stem locations. It adds therapeutic effect without adding significant toxicity. In order to improve its contribution, it is essential to find new drugs with more penetration in order to destroy tumor cells more deeply at resection margins.

51 Keywords: photodymamic therapy; brain tumors; neurosurgery

ip t

52 53

55

cr

54

All the authors above mentioned declare that there are no conflicts of interest

us

56

an

57 58

M

59 60

62

te

d

61

INTRODUCTION

64

Gliomas account for approximately 40~50% of all intracranial malignancies. The prognosis

65

of patients with malignant glioma is dismal, especially those harboring a glioblastoma

66

(GBM), with a median survival of less than 2 years after the initial diagnosis1, 2. Over the

67

years and despite all attempts to improve the outcome with surgery, radiotherapy and

68

chemotherapy, there has been only very little improvement. Thus new treatment modalities

69

are needed.

70

Survival of patients suffering from malignant brain tumors is highly dependent on the

71

extent of surgical resection, but the completeness of tumor resection has to be maximized

72

without affecting critical brain structure and function. As most recurrences occur within 2

73

cm of the original tumor site1, more aggressive local therapies are necessary to eradicate

74

tumor cells that invade adjacent normal brain tissue. Standard postoperative treatment is a

75

combination of radio and chemotherapy including temozolomide2, 3.

Ac ce p

63

3 Page 3 of 17

Photodynamic therapy (PDT) constitutes a treatment modality that combines a

77

photosensitizing agent with exposure to laser light in order to elicit phototoxic reactions

78

that selectively destroy tumor cells and spare normal cells4.

79

PDT involves the administration of a photosensitizer and the illumination of the target with

80

light of a wavelength corresponding to the absorption peak of the administered drug. The

81

absorption of photon energy by the sensitizer induces a photochemical reaction that results

82

in cellular damage leading to cell death4. Cancer cells retain more photosensitizer inside

83

them than surrounding normal cells after a specified amount of time has passed, causing a

84

relative selectivity regarding this treatment5. Additional advantages of PDT are that it can

85

induce apoptosis in cancer cells, with no effects in the extra-cellular matrix that remains

86

forming a scaffold for the surrounding normal tissue to advance over and it has vascular

87

mediated effects that contribute to tumor control4-6.

88

PDT is a local treatment modality. Its application can be repeated more than once to the

89

same area without accumulative effects7, 8. It does not negatively affect other treatment

90

modalities, such as radiation or chemotherapy, leaving further treatment options open.

91

All the characteristics described, in addition to the initial favorable experiences

92

communicated by other groups4, 5, prompted us to perform a feasibility study of PDT in

93

brain tumors. Initially porfimer sodium (Photofrin™, Pinnacle Biologics, Bannockburn IL,

94

USA) was chosen4, 5. When second-generation photosensitizers were ready to use, we

95

switched to temoporfin (meta-tetrahydroxyphenylchlorin –mTHPC-, Foscan™ Biolitec,

96

Jena, Germany)9-11. mTHPC has a deeper penetration in brain tissue and it is cleared from

97

the skin much faster than porfimer sodium.

99

cr

us

an

M

d

te

Ac ce p

98

ip t

76

PATIENTS AND METHODS

100

Study Subjects

101

Patients diagnosed with primary brain tumors and neither contraindications for general

102

anesthesia nor brain surgery were included. After death of two patients with thalamic

103

tumors, protocol was modified, excluding patients with thalamic and brain stem locations. 4 Page 4 of 17

Study was reviewed and approved by the Hospital Ethics Committee. Written informed

105

consent and Compassionate Use approval from Spanish Health Ministry were obtained for

106

each patient according to Spanish regulation.

107

PDT Therapy

108

Forty eight hours before the surgical procedure, porfimer sodium was administered at a

109

dose of 2 mg/kg body weight, through an intravenous drip line. The infusion lasted over 30

110

minutes. Photoactivation of porfimer was obtained during surgery by exposure of the

111

residual cavity to a non-thermal light at a wavelength of 630 nm, provided by a Ceralas

112

PDT 630 Diode Laser (CeramOptec GmbH, Bonn, Germany). The light dose administered

113

in this study was 75 J/cm2.

114

mTHPC was administered 96 hours before surgery, at 0.15 mg/kg body weight, into a thick

115

vein of the arm for 6 minutes. Photoactivation of mTHPC was obtained during surgery by

116

exposure of the surgical cavity to a non-thermal light at a wavelength of 652 nm provided

117

by a laser Ceralas PDT 652 (CeramOptec, Bonn, Germany). The light dose administered

118

was 20 J/cm2.

119

Usually post-excision cavities vary in size and are irregular in shape. The Neurosurgeon

120

calculates the amount of light to be delivered to the treatment site with the assistance of

121

both Radiotherapy and Medical Physics specialists. Two optical fibers were used, alone or

122

in combination: one designed to produce an uniform circular field, suitable for illumination

123

of flat surfaces and one diffuser fiber for obtaining a spherical field. Personalized fields

124

were calculated for every patient with the aim of avoiding dark zones and overdosed ones.

125

Once in place, the fibers were maintained in the adequate position by means of a

126

mechanical arm.

127

According to the protocol and the recommendations of the manufacturers, an illustrated

128

booklet explaining detailed written guidelines for light exposure was provided to the

129

patient, families and referring physicians, making them fully aware about the importance of

130

complying with the light exposure requirements in PDT.

131

Surgical procedure

Ac ce p

te

d

M

an

us

cr

ip t

104

5 Page 5 of 17

All pre-operative studies were performed before administering the photosensitizing agent.

133

The surgical procedure took place 48 hours after porfimer sodium administration and 96

134

hours after mTHPC administration, respectively.

135

Craniotomy with radical tumor resection under microscopic guidance was performed. Once

136

the surgical removal of the tumor was finished, a laser light was applied to the tumor bed to

137

destroy the remaining tumor cells invading the normal looking brain tissue. Light was

138

delivered with a laser at 630 nm, 75 J/cm2 in porfimer sodium patients and at 652 nm, 20

139

J/cm2 in the mTHPC ones. While the laser light was applied the surgical cavity was cooled

140

with a continuous infusion of Intralipid™ (Fresenius Kabi, Barcelona, Spain) at a

141

concentration of 2%, diluted in saline serum. Standard procedures for safe laser use were

142

strictly followed. In the operating room no direct light was used and the areas of the patient

143

not exposed in the surgical procedure were covered with thick blankets. In the surgical

144

field, the non-tumor areas were covered with moist gauzes. Afterwards the wound was

145

closed in the usual manner. Once in the ICU, patients were kept in a special room with no

146

natural sunlight.

147

Post-surgery procedure

148

Patients left the hospital usually 5-7 days post-op, at night, after sunset. Once at home,

149

sunlight was avoided until 4 weeks (porfimer sodium) or 2 weeks (mTHPC) had elapsed

150

since the administration of the photosensitizing agent. Patients’ individual sensitivity to

151

light was tested. In any case patients were instructed to observe precautions to avoid intense

152

sunlight exposure for the first 7 days.

153

Post-operatively, patients received temozolomide (TMZ)-based chemotherapy and

154

radiotherapy (RT, 60 Gy) according to Stupp protocol2, 3. In case of progression to previous

155

TMZ and/or RT, they received intraarterial cis-platinum and bevacizumab. Radiosurgery

156

was considered for small recurrences.

157

Patients were evaluated with physical examination, blood test including hemogram and

158

basic biochemistry and MRI at 1 and 3 months and then at 3 months interval until relapse

159

or death.

160

Statistical analysis

Ac ce p

te

d

M

an

us

cr

ip t

132

6 Page 6 of 17

161

Data are reported as means and 95% confidence interval (CI). All statistical tests were

162

performed using the SPSS 17.0 software (SPSS, Chicago, IL, USA). The survival data were

163

analyzed by Kaplan-Meier method.

ip t

164

RESULTS

166

From May 2000 to December 2010, 41 patients (27 male, 14 female) with a median age of

167

49 years (range 13 to 70) diagnosed of primary brain tumors were treated with PDT.

168

Histologies were GBM (n=20), anaplastic astrocytoma (AA, n=10), low grade astrocytoma

169

(LGG, n=4), oligodendroglioma (OD, n=7, 4 anaplastic). In 7 patients PDT was repeated at

170

the time of the relapse, and for the purpose of analysis of progression-free survival (PFS)

171

and overall survival (OS) they have been considered as separate procedures. In 22 episodes

172

PDT was part of the initial treatment and in 26 episodes was part of the treatment at relapse.

173

Therapy side effects

174

Two patients with thalamic tumors treated at the initial phase of the study died in early

175

post-op period. A 40-y old male diagnosed with GBM of left thalamic region received PTD

176

with Photofrin after a complete resection. He presented massive edema at the end of the

177

procedure, refractory to aggressive antiedema measures. The other, a 40-y old male

178

diagnosed with AA of the left thalamic region, was operated, leaving a plaque of residual

179

tumor close to the internal capsule. He received PTD with Photofrin. Two days later he

180

presented massive and refractory edema. We decided to add thalamic or brain stem location

181

as absolute exclusion criteria. Otherwise, no other cases of massive edema were observed.

182

Mild postoperative brain edema was adequately controlled with corticosteroid treatment.

183

Neurologic post-operative deficits occurred in 5 patients (4 hemiparesia, 1 hydrocephalus, 1

184

ptosis). Three patients with hemiparesia recovered in 3 to 17 days with dexametasone and

185

mannitol; the fourth continues alive but with residual right hemiparesia, 23 months after

186

surgery. The hydrocephalus required surgical correction with brain-abdominal derivation.

187

Most patients complained of moderate pain in the operated area in the first days after

188

treatment, which could be adequately managed with common analgesics.

Ac ce p

te

d

M

an

us

cr

165

7 Page 7 of 17

One scalp burn requiring plastic surgery with cutaneous graft was observed. Other

190

cutaneous toxicities were one case of blisters on the forearm and other of erythema in

191

forearm, caused by a negligent observation of specific protective measures about light

192

exposure. A case of burn on the thumb fingernail was also observed, induced by the

193

pulsoxymeter in ICU. Since then, the pulsoxymeter was changed of finger every hour and

194

not used when the patient was fully awake. Slight dermatitis along the course of the vein

195

used for the injection of the photosensitizer was almost universally observed. Dermatologic

196

complications were usually well managed with conservative treatment and resolved without

197

sequealae.

198

Survival

199

Median PFS observed was 10 months for GBM (95% confidence interval 5.7-14.3), 26

200

months for AA (95% CI 4.5-47.5), and 43 months for OD (95% CI 4.5-47.5). Median OS

201

was 9 months for GBM (95% CI 2.3-15.7), 20 months for AA (95% CI 0.0-59) and 50

202

months for OD (95% CI 32.5-67.5). The apparent discrepancy between PFS and OS data is

203

due to 1 patient with GBM and 2 with AA not censored for PFS because they die from

204

causes other than tumor progression (1 pneumonia, 1 cardiomiopathy, 1 sepsis). Median OS

205

since first diagnosis was 17 months for GBM (95% CI 15.2-17.8), 66 months for AA (95%

206

CI 2.9-129.1) and 122 months for OD (95% CI 116.1-127.8) (see Figure 1).

207

Four patients are alive at the moment without evidence of active disease: one patient with

208

OD, treated with porfimer sodium at the third relapse and with mTHPC at the fourth relapse

209

(238 months since initial diagnosis), one with LGG treated with porfimer sodium in the

210

initial surgery (152 months since first diagnosis), one with OD, treated with mTHPC at the

211

third relapse after two previous surgeries (142 months since initial diagnosis), and the last

212

one with LGG, treated with porfimer sodium in the initial surgery and with mTHPC at

213

relapse (132 months since first diagnosis).

214

In several cases of high grade glioma a particular pattern of progression was observed. It

215

consisted in tumor growth in points of the surgical bed where a non-optimal exposure to the

216

laser light was suspected considering its irregular shape as well as recurrence beyond the

217

edges of the surgical cavity and/or distant recurrence including the contralateral

218

hemisphere.

Ac ce p

te

d

M

an

us

cr

ip t

189

8 Page 8 of 17

In 22 episodes PDT was part of initial treatment of primary brain tumors and in 26 episodes

220

was part of the treatment at relapse. Twenty one procedures were performed using porfimer

221

sodium and 27 using mTHPC. Comparisons in efficacy between photosensitizers have not

222

been possible due to the heterogeneity of the series and the small number of patients in each

223

group to draw definitive conclusions.

ip t

219

cr

224

DISCUSION

226

Local invasion is characteristic of glioma growth and the most frequent cause of recurrence.

227

Around 90% of recurrences diagnosed after initial surgery are localized in a 2-cm area

228

surrounding the primary tumor bed1,

229

critical for the prevention of glioma recurrence.

230

Adjuvant RT, alone or in combination with chemotherapy, has been administered after

231

surgery with the aim of killing any tumor cell infiltrating normal brain. Multiple trials with

232

different agents and schedules were published with no clear benefit. In 2005, Stupp and

233

cols. reported the final results of a randomized trial, showing that the addition of TMZ to

234

RT as initial adjuvant treatment prolonged survival of patients with GBM. Five hundred

235

seventy-three patients were randomly assigned to either standard RT alone or RT and

236

concomitant and maintenance administration of TMZ (TMZ/RT). Patients treated with

237

TMZ/RT had a median survival time of 15 months, compared with 12 months for the

238

patients initially treated with RT alone. The 2-year survival rate was 26% for the TMZ/RT

239

group compared with only 10% in the RT group2, 3.

240

Despite these encouraging results, chemo and RT continue to present important limitations.

241

RT induces damage and long term effects on the normal brain tissue, a tribute frequently

242

paid in the attempt to control tumor growth. New modalities as image modulated RT

243

(IMRT) or 3D conformation permit a more selective radiation delivery to the critical areas.

244

In selected patients, a second irradiation is feasible, but still far from a dose that could be

245

considered curative.

an

. Therefore, removal of residual cancer cells is

Ac ce p

te

d

M

12

us

225

9 Page 9 of 17

Some encouraging results were published with high dose chemotherapy in the context of

247

multidisciplinary programs and selected patients, but, in general, no significant advances in

248

chemotherapy of gliomas have been reported after TMZ13.

249

The study AVAglio has demonstrated an improvement in PFS without significant changes

250

in OS with bevacizumab, an antiangiogenic drug. It has shown also an improvement in

251

quality of life reflected in higher time to neurological deterioration and low corticoids

252

consumption in comparison with non-bevacizumab treated patients. On the other hand,

253

antiangiogenics interfere with the normal healing of the surgical wounds. Definitive role of

254

bevacizumab in gliomas has to be defined14.

255

In any case, the current survival rates for malignant brain gliomas treated with surgery,

256

radio and chemotherapy has not significantly changed over the years, showing that more

257

effective treatment strategies need to be explored.

258

Use of 5-amino-delta-levulinic acid or other photoactive agents has been postulated for

259

increasing the radicality of the surgical removal, helping to detect residual tumor by its

260

luminescence, otherwise not observed by the naked eye7,

261

radical removal is not always possible in the brain as the surgical excision of small amount

262

of tissue in relevant brain areas can result in significant morbidity.

263

PDT has been applied with great success in many areas of medicine. Its first application to

264

brain tumors dates back to the 1988 Kaye's team series17. This group treated 56 brain tumor

265

cases with a mixed pathology and saw no increase in permanent neurological deficits with

266

no technical complexity and with a promising increase in the survival rate. They later

267

updated their series confirming initial observations18, 19. Other reports from Canada20 and

268

those from Eljamel et al.8, 21-23 in recent years have not only reproduced initial results but

269

also have explored modifications in technique in an attempt to improve final results. New

270

promising porfirin derivatives are being currently evaluated for clinical use24.

271

Data from this study confirm the feasibility of adding PDT to the standard therapy of

272

malignant brain tumors. The results found in this series are not inferior to those found in

273

literature, in spite of the particular characteristics of the patients. Nineteen of the 41 patients

274

were treated at relapse, after one or more surgeries and chemo/RT. The number of patients

275

operated with recurrences makes this series more valuable.

8, 15, 16

.Unfortunately such a

Ac ce p

te

d

M

an

us

cr

ip t

246

10 Page 10 of 17

Some LGG can be controlled with standard treatment. Nevertheless, the pre-op diagnosis of

277

a LGG is frequently based in stereotactic biopsies and/or radiologic findings, and it has to

278

be remarked that an unequivocal diagnosis is obtained only after tumor removal when the

279

anatomical location permits it. Mixed tumors, unusual radiologic findings and relapses are

280

not uncommon and these were the reasons for including LGG in the study25.

281

Local control of the disease has been satisfactory, particularly in LGG. Recurrences in

282

GBM often happened at distant sites from the PDT treated surgical bed. In some cases,

283

including two of the patients alive at this moment, longer survivals were obtained after re-

284

operation with PDT, suggesting that PDT can be repeated with no accumulative effects.

285

In young patients the possibility of treating the tumor without RT is particularly useful,

286

since it avoids the potential for radiation-induced tumors later in life, and prevents the long-

287

term sequelae after RT. That is the case of one of our patients treated when he was 13 years

288

old for a recurrent OD. In spite of not receiving RT is still free of recurrence eight years

289

later.

290

Both reported deaths were due to strong post-op edema in thalamic region. Patients with

291

thalamic and brain stem tumors should be discarded for PDT until more effective methods

292

for controlling immediate post-op edema are available.

293

mTHPC infusion-related local pain has been repeatedly reported6. In our series we found

294

essential that the intravenous injection be carried out slowly (the manufacturer's

295

recommendation is over 6 minutes) and in a thick caliber vein of the forearm.

296

Photosensitivity skin reactions are avoidable. Minor skin photosensitivity reactions due to

297

exposure to natural sunlight might be completely avoided by strict compliance of the light

298

exposure recommendations. In our series, none was of clinical relevance except for the

299

scalp burn requiring plastic surgery. There are other concerns such as photosensitizer

300

extravasation while being injected and subungual burn due to pulse oxymeter, that need

301

careful attention of the caring team to avoid them.

302

Slight dermatitis along the course of the vein used for the injection of the photosensitizer

303

was almost universally observed. The most probable origin is microextravasation, causing

304

retention of the drug in perivascular area.

Ac ce p

te

d

M

an

us

cr

ip t

276

11 Page 11 of 17

Although the present and some previous PDT studies are based on a relatively small

306

number of patients and in inhomogeneous samples, they suggest an encouraging benefit for

307

patients with brain tumors. This study confirms that PDT can be considered as an

308

adjunctive to surgery and/or RT and chemotherapy in the treatment of brain tumors,

309

excluding those patients with thalamic or brain stem locations.

ip t

305

310

REFERENCES

313

1.

314

2.

an

468.

316 317

Burton EC, Prados MD. Malignant gliomas. Curr Treat Options Oncol 2000;1:459-

Stupp R, Mason WP, van den Bent MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–996.

3.

M

315

us

312

cr

311

Stupp R, Hegi ME, van den Bent MJ et al. Changing paradigms – An update on the multidisciplinary management of malignant glioma. Oncologist 2006;11:165-180.

319

4. Dougherty TJ, Gomer CJ, Henderson BW et al. Photodynamic therapy. J Natl Cancer

5.

322 323

Laser Med Surg 1996;14:239-244. 6.

324 325

Biel MA. Photodynamic therapy and the treatment of head and neck cancers. J Clin

Ac ce p

321

Inst 1998;90:889-905.

te

320

d

318

Kostron H, Obwegeser A, Jakober R. Photodynamic therapy in neurosurgery: a review. J Photochem Photobiol B 1998;36:157-168.

7.

Zilidis G, Aziz F, Telara S, Eljamel MS, Fluorescence image-guided surgery and

326

repetitive photodynamic therapy in brain metastatic malignant melanoma. Photodiag

327

Photodyn Ther 2008;5:264-266.

328

8. Eljamel MS, Goodman C, Moseley H. ALA and Photofrin™ fluorescence-guided

329

resection and repetitive PDT in glioblastoma multiforme: a single centre Phase III

330

randomised controlled trial. Lasers Med Sci 2008; 23:361-367.

331 332

9. Cramers P, Ruevekamp M, Oppelaar H et al. Foscan uptake and tissue distribution in relation to phodynamic efficacy. Br J Cancer 2003;88:283-290. 12 Page 12 of 17

333

10. Copper MP, Tan IB, Oppelaar H et al. Meta-tetra(hydroxyphenyl)chlorin photodynamic

334

therapy in early-stage squamous cell carcinoma of the head and neck. Arch Otolaryngol

335

Head Neck Surg 2003;129:709–711. 11. D’Cruz AK, Robinson MH, Biel MA. mTHPC-mediated photodynamic therapy in

337

patients with advanced incurable head and neck cancer: a multicenter study of 128

338

patients. Head Neck 2004;26:232-240.

cr

340

12. Giese A, Westphal M. Glioma invasion in the central nervous system. Neurosurgery 1996;39:235-250.

us

339

ip t

336

13. Fernández-Hidalgo OA, Vanaclocha V, Vieitez JM et al. High dose BCNU and

342

autologous progenitor cell transplantation given with intra-arterial cisplatinum and

343

simultaneous radiotherapy in the treatment of high-grade gliomas: benefit for selected

344

patients. Bone Marrow Transplantation 1996;18:143-149.

an

341

14. Henriksson R, Mason W, Chinot O, Wick W on behalf of the AVAglio Steering

346

Committee. Bevacizumab in Glioblastoma: Another Perspective. The ASCO Post 2013;

347

July 10, Volume 4, Issue 11.

349

d

15. Eljamel MS. Fluorescence image guided surgery of brain tumours: Explained step by

te

348

M

345

step. Photodiag Photodyn Ther 2008;5:260-263. 16. Eljamel MS. Photodynamic assisted surgical resection and treatment of malignant brain

351

tumours technique, technology and clinical application. Photodiagn Photodyn Ther

352

2004;1:93-98.

353 354 355 356 357 358 359 360

Ac ce p

350

17. Kaye AH, Morystyn G, Apuzzo M. Photoradiation therapy and its potential in the management of neurological tumors: a review. J. Neurosurg 1988;69:1-14. 18. Stylli SS, Kaye AH, MacGregor L et al. Photodynamic therapy of high grade glioma long term survival. J Clin Neurosci 2005;12:389-98. 19. Stylli SS, Kaye AH. Photodynamic therapy of cerebral glioma - a review. Part II clinical studies. J Clin Neurosci 2006;13:709-17. 20. Muller PJ, Wilson BC. Photodynamic therapy of malignant brain tumors. Lasers Med Sci 1990;5:245-251. 13 Page 13 of 17

361

21. Eljamel MS. New light on the brain: the role of photosensitising agents and laser light

362

in the management of invasive intracranial tumours. Technol Cancer Res Treat

363

2003;2:303- 309.

367 368

ip t

366

Photodiagn Photodyn Ther 2005; 1: 303- 310.

23. Moseley H, McLean C, Hockaday S, Eljamel S. In vitro light distributions from intracranial PDT balloons. Photodiag Photodyn Ther 2007;4:213- 220.

cr

365

22. Eljamel MS. Brain PDD and PDT unlocking the mystery of malignant gliomas.

24. Hiramatsu R, Kawabata S, Miyatake SI et al. Application of a novel boronated

us

364

porphyrin (H2OCP) as a dual sensitizer for both PDT and BNCT. Lasers Surg Med

370

2011;43:52-58.

371

an

369

25. Grier JT, Batchelor T. Low-grade gliomas in adults. The Oncologist 2006;11:681-693.

372

Ac ce p

te

d

M

373

14 Page 14 of 17

373

Figure 1

1,0

0,8

0,8

0,6

0,6

0,0

0,0

0

12

24

36

48

60

72

84

96

108

120

0

12

24

36

48

60

72

84

MONTHS

OS

an

PFS

M

1,0

d

0,6

0,4

0,2

0,0

380

24

36

48

60

72

84

96

108 120 132 144 156 168 180

Ac ce p

379

12

te

S U R V IV A L

0,8

378

108 120 132 144 156 168 180

MONTHS

376

377

96

us

0,2

0,2

375

0,4

cr

0,4

ip t

1,0

S U R V IV A L

S U R V IV A L

374

MONTHS

OS since diagnosis

16 Page 15 of 17

380

LEGEND

381

Figure 1

382

Kaplan Meier curves corresponding to PFS, OS and OS since diagnosis, for GBM (••••), AA (•−•−•), OD

383

(――) and LGG (

)

ip t

384

Ac ce p

te

d

M

an

us

cr

385

17 Page 16 of 17

385

Picture 1. A general view of the surgical theater during the PTD procedure

386 387

Picture 2. A detail of the surgical bed during illumination phase, showing cooling with Intralipid solution

388

Picture 3. Register formulary for the procedure parameters

Ac ce p

te

d

M

an

us

cr

ip t

389

18 Page 17 of 17

Photodynamic therapy in the treatment of brain tumours. A feasibility study.

Photodynamic therapy (PDT) constitutes a treatment modality that combines a photosensitizing agent with exposure to laser light in order to elicit pho...
166KB Sizes 3 Downloads 7 Views