• Non ci sono risultati.

11.6 Translational Research in Radiation Oncology of Lung Cancer

N/A
N/A
Protected

Academic year: 2022

Condividi "11.6 Translational Research in Radiation Oncology of Lung Cancer"

Copied!
13
0
0

Testo completo

(1)

11.6 Translational Research in Radiation Oncology of Lung Cancer

Yuhchyau Chen

Y. Chen, PhD, MD

Associate Professor, Director of Clinical Investigation, Department of Radiation Oncology, 601 Elmwood Ave., Box 647, Rochester, NY 14642, USA

CONTENTS

11.6.1 Introduction 481

11.6.2 Optimizing Paclitaxel Chemoradiation Sensitizing Strategy in Stage III

Non-Small Cell Lung Cancer: A Phase I/II Study Based on Pre-Clinical Research 481 11.6.2.1 Paclitaxel Chemoradiation for NSCLC 482 11.6.2.2 Pharmacokinetic Consideration

of Paclitaxel for Radiosensitization 483 11.6.2.3 Paclitaxel Cell Cycle Effect on Human

Cancer Cell Lines 484 11.6.2.4 Paclitaxel Apoptotic Effect 484 11.6.2.5 Schedule-Dependent Radiosensitizing Effect In Vitro 484

11.6.2.6 Clinical Protocol of Pulsed Paclitaxel and Radiation 485

11.6.2.7 Tumor Response, In-Field Tumor Control, and Survival 485

11.6.2.8 Toxicity 486

11.6.2.9 Future Directions in Chemopotentiation of Radiotherapy for Stage III NSCLC 487 11.6.3 Cytokine Markers for Radiation

Pulmonary Injury 487 11.6.3.1 Radiation Lung Injury 487

11.6.3.2 Cytokine and Radiation Lung Injury 488 11.6.3.3 Infl ammatory Cytokines IL-1α and IL-6 488 11.6.3.4 Chemokines and Cell Adhesion Molecules 489 11.6.3.5 Fibrotic Cytokine: Transforming

Growth Factor β 490 11.6.4 Conclusions 490 References 491

11.6.1 Introduction

Although radiation is an essential modality for can- cer therapy because of its powerful DNA-damaging effects, those same effects unfortunately cause si- multaneous injury to patients’ normal tissues. Thus the success of radiotherapy relies on the fi ne bal- ance between eradicating cancer cells and minimiz-

ing injury to normal tissues. In lung cancer research and treatment, basic biomedical research and clini- cal studies have fl ourished in the past two decades, but lung cancer remains the most deadly cancer of all types. Despite the explosion of discoveries that have elucidated our understanding of the molecular mechanisms and regulatory signals involved in can- cer progression, transferring knowledge into practi- cal advances for treating lung cancer and preventing side effects from cytotoxic therapy has been a more deliberate process. The translation of knowledge gained from the laboratory into improving treatment outcomes will serve as the vital bridge between sci- entifi c discovery and the welfare of patients and will offer hope for further improvement of lung cancer therapy.

Because the scope of this chapter is limited, it will focus on two areas of translational research in radiation oncology for lung cancer treatment: 1) the attempt to potentiate radiation effects by a unique radiosensitizing strategy that optimally integrates paclitaxel and radiation to enhance radiation’s local effects in treating locally advanced non-small cell lung cancer (NSCLC), and 2) the cytokines involved in radiation lung injury that lead to radiation pneu- monitis and progressive fi brosis.

11.6.2

Optimizing Paclitaxel Chemoradiation Sensitizing Strategy in Stage III Non-Small Cell Lung Cancer: A Phase I/II Study Based on Pre-Clinical Research

The treatment of locally advanced, stage III NSCLC remains a challenging task for oncologists because of lack of local disease control and high rates of distant disease failures. Although aggressive combination chemoradiation therapy is now the new standard of treatment for stage III NSCLC, 5-year survival rates are only in the range of 5%–25%, and local failure rates have been as high as 55%–85% (Dillman et

(2)

al. 1990; Sause et al. 2000; Schaake-Koning et al.

1992; Arriagada et al. 1991; Morton et al. 1991).

In addition, due to aggravated toxicity from therapy, combined modality treatment is offered to a limited population of patients in good clinical performance status only. Both the optimal regimen and sequence of chemoradiation therapy for stage III NSCLC re- main unclear.

At least two randomized studies have demon- strated that concurrent chemoradiation is more ef- fective than sequential chemotherapy followed by radiation for the treatment of stage III NSCLC, sup- porting the radiosensitization mechanism in im- proving survival of lung cancer patients (Furuse et al. 1999; Curran et al. 2000). The phase III ran- domized study conducted by the Radiation Therapy Oncology Group (RTOG) for stage III NSCLC repre- sents a good example of the dilemma in trading tox- icity for effi cacy. RTOG 94–10 compared three arms of chemoradiation treatments: sequential chemora- diation vs. concurrent chemotherapy and once-daily (QD) radiation vs. concurrent chemotherapy and twice-daily (BID) radiation. The combined chemora- diation strategy resulted in a modest survival gain in the concurrent chemoradiation arm, with a median survival of 17 months for the concurrent QD arm vs.

14.6 months for the sequential chemoradiation arm (P =.038), and a median survival of 15.6 months for the concurrent BID arm. Most notable was a dra- matic increase in grade 3/4 acute nonhematologic toxicities such as esophagitis, pneumonitis, and oth- ers, observed in 48% of the concurrent QD arm, 30%

of the sequential arm, and 62% of the concurrent BID arm (Curran et al. 2000). The observed increase in normal tissue toxicity in concurrent chemoradiation treatment supports the notion that while concurrent chemoradiation enhances radiation tumoricidal ef- fects, it simultaneously enhances injuries to normal tissues. As cancer therapy continues to explore newer agents in combination with radiation, balancing the therapeutic effect and treatment-related toxicity will also continue to challenge oncologists.

11.6.2.1

Paclitaxel Chemoradiation for NSCLC

Despite the convincing evidence from randomized studies of stage III NSCLC that chemoradiation is su- perior to radiation alone, the optimal chemotherapeu- tic agent(s), dosing schedule, sequence, and timing of chemoradiation to achieve the best therapeutic gain remain unknown. Because of lack of effective therapy

for stage III NSCLC, newer agents including the 3rd- generation chemotherapeutic agents have been tested with the intent to improve the effi cacy of combination chemoradiation for stage III NSCLC over cisplatin- based chemoradiation (Chen and Okunieff 2004).

Among the 3rd-generation chemotherapeutic agents, paclitaxel has been the favorite chemotherapeutic agent investigated in many phase I/II clinical studies due to its putative radiosensitizing effect. Paclitaxel, as a single agent, has demonstrated response rates of 21–24% in the metastatic setting (stage IV) of NSCLC (Murphy et al. 1993). It is theoretically the ideal ra- diosensitizer due to its cytokinetic stabilization of the spindle microtubule resulting in arresting cells in the G2/M phase of the cell cycle, which is the most radiosensitive phase of the cell cycle (Sinclair and Morton 1966; Schiff and Horwitz 1980; Parness and Horwitz 1981; Manfredi et al. 1982; Kumar 1981; Rowinsky et al. 1988). The cytotoxic effect is at- tributed to tumor apoptosis after treatment with pacli- taxel (Milas et al. 1995, Milross et al. 1996; Jordan et al. 1996; Pukkinen et al. 1996). For the interaction with radiation treatment, in vitro studies have also demon- strated the radiosensitizing effects of paclitaxel in a variety of cancer cell lines, including cervical cancer, ovarian cancer, astrocytoma, melanoma, pancreatic carcinoma, breast cancer, colon cancer, lung cancer, prostate adenocarcinoma, and others (Tishler et al.

1992; Steren et al. 1993; Geard et al. 1993; Rodriguez et al. 1995; Lokeshwar et al. 1995; Elomaa et al. 1995;

Choy et al. 1993; Jordan et al. 1996; Pukkinen et al.

1996; Hornback et al. 1994). In addition to cell cycle and apoptotic effects, paclitaxel offers antineoplastic potential through other mechanisms such as enhanc- ing tumor reoxygenation, thereby reducing the resis- tance of hypoxic cells to radiation and chemotherapy (Milas et al. 1995a). Furthermore, paclitaxel exhibits antiangiogenic effects on tumor vasculature by caus- ing apoptosis of endothelial cells as well (Grant et al.

2003). In the clinical setting, there have been several different dose schedules of paclitaxel-based chemora- diation regimens reported for treating stage III NSCLC (Choy et al. 1994; Lau et al. 1997; Chen et al. 2003;

Rathmann et al. 1999; Kirkbride et al. 1999; Brodin et al. 2000; Havemann et al. 1995; Rosenthal et al.

2000), but the optimal dosing schedule in integrating paclitaxel with radiation remains to be defi ned.

Because paclitaxel is a cell-cycle-specifi c agent, its effect can theoretically be optimized by timing radia- tion to allow for radiation injury at the G2/M phase of cancer cells. Because normal tissue and cancer cells have different growth kinetics, minimizing normal tissue injury is also possible if information from pre-

(3)

clinical research and pharmacokinetics are taken into consideration in the design of a clinical study. Here a phase I/II clinical study will be presented to illustrate the application of preclinical research on lung cancer cell lines and the pharmacokinetic characteristics of paclitaxel infusions to the design of the clinical study.

The hypothesis is that maximum tumor control and minimal normal tissue injury can be achieved by strategically optimizing the timing of daily irradia- tion and low-dose paclitaxel treatment.

11.6.2.2

Pharmacokinetic Consideration of Paclitaxel for Radiosensitization

Understanding the pharmacokinetics of paclitaxel is essential for optimizing the timing of paclitaxel chemotherapy and radiation treatments to enhance radiation effects and reduce normal tissue side ef- fects. The pharmacokinetics of paclitaxel infusion are unique in that not only does the drug concentration infl uence the pharmacokinetics of the drug, but also the infusion time signifi cantly infl uences the phar- macokinetics of plasma concentration. Furthermore, bone marrow toxicity is directly related to the infu- sion time and drug concentration. Pharmacokinetic studies have reported a nonlinear disposition of pa- clitaxel in humans. In the reported clinical trials, the infusion time of paclitaxel has varied signifi cantly:

1 h, 3 h, 24 h, or continuous. Gianni et al. (1995) com-

pared the nonlinear disposition of paclitaxel with different drug concentrations and infusion times.

As demonstrated in Fig. 11.6.1a, the shorter infusion time of 3 h resulted in a fairly rapid increase to a high peak plasma level at 2–3 h after infusion, which was followed by a rapid decline of plasma concentrations.

The longer infusion time of 24 h was associated with a rise in plasma level to a moderately high plateau level for 24 h, followed by a slower decline of the drug concentrations.

As described by the same investigators, the de- crease of absolute neutrophil counts from paclitaxel treatment was directly associated with the “dura- tion” of plasma drug concentration above 0.05 µM (Fig. 11.6.1b). Thus, the long plateau of plasma con- centration ≥0.05 µM from a 24-h infusion was as- sociated with more myelotoxicity, and the shorter infusion time of 3 h was associated with less myelo- toxicity despite the higher initial peak plasma level.

In the design of a clinical study when neutropenia is a concern and when radiosensitization is the primary interest, the choice of the shorter infusion time will be the logical one.

11.6.2.3

Paclitaxel Cell Cycle Eff ect on Human Cancer Cell Lines

The cell cycle effect of paclitaxel on human cancer cell lines was investigated in cell lines A431, A549, and

Fig. 11.6.1 a Plasma paclitaxel concentration vs. time profi les of representative patients who received the drug at various indi- cated doses and infusion durations. Symbols represent actual measured plasma paclitaxel concentrations and lines represent model fi t curves. b Pharmacokinetic/pharmacodynamic relationship between duration spent at a plasma paclitaxel concentra- tion ≥0.05µM/l and percentage reduction in absolute neutrophil count in the fi rst course of therapy. Symbols represent individu- als treated at different doses and schedules. Curve depicts the sigmoid Emax model fi t to the data. The broken portion of the curve represents that region for which data were not available. Adapted from Gianni et al. (1995). Reprinted with permission from the American Society of Clinical Oncology

a b

Time (h) Time plasma [PACLITAXEL] ≥ 0.05 µM (h)

Plasma [PACLITAXEL] (µM) % Decrease in ANC

(4)

NCI-H520. A431 is a human epidermoid cancer cell line; A549 and NCI-H520 are both human lung can- cer cell lines. Fig. 11.6.2a demonstrates the cell cycle progression of cell line A431 after a 3 h treatment with 1µM paclitaxel. G2/M accumulation was ap- preciated at approximately 4 h after drug treatment.

Further cell cycle progression to the G2/M phase was observed and was followed by a dynamic redistribu- tion of cell cycle to return to the baseline by 48 h.

In an attempt to sustain the G2/M cell cycle arrest for maximal radiosensitizing effect, an in vitro study was conducted by pulsing paclitaxel every 48 h and analyzing the cell cycle effect 24 h later. Fig. 11.6.2b shows that by pulsing paclitaxel on alternate days using one-third of the concentration (0.33 µM), there was sustained cell cycle arrest in the G2/M phase for 6 days. Sustained G2/M cell cycle arrest by pulsing paclitaxel at lower doses was observed in all three cancer cell lines and supported the application of this schedule to the design of a clinical trial (Chen et al. 2003a).

11.6.2.4

Paclitaxel Apoptotic Eff ect

In the current study, cells were treated with paclitaxel for 3 h and assayed by the Tunnel assay for apopto- sis. As shown in Fig. 11.6.3, although the percent-

age of apoptotic cells varied among these three cell lines, common observations were made in that higher doses (600 nM and 2 µM) of paclitaxel caused more apoptosis than the lowest dose (200 nM). It was also observed that more apoptosis occurred at 48 h after drug treatment than at 24 h. The timing of maximal paclitaxel apoptotic effect also supports pulsing pa- clitaxel treatment every 48 h (Chen et al. 2003a).

11.6.2.5

Schedule-Dependent Radiosensitizing Eff ect In Vitro

In preliminary studies, Keng et al. (2000) and Chen et al. (2001) investigated the schedule dependence of paclitaxel interaction with radiation in three lung cancer cell lines. Cells were treated in the cul- ture with either 50 nM or 100 nM of paclitaxel for 3 h. The control received no paclitaxel. Radiation at various doses was delivered either immediately after the 3-h drug treatment (labeled as 3 h) or 21 h after drug treatment (labeled as 24 h). The clono- genic survival was assayed 2 weeks after radiation.

The survival curves showed that delaying radia- tion resulted in fewer survival clones. When other cell lines were tested, it appeared that paclitaxel chemopotentiation of radiation was often through the sub-additive mechanism rather than an addi-

Fig. 11.6.2 a Human epidermoid cell line A431 culture was exposed to 1.0 µM paclitaxel for 3 h.

After 3 h the drug-containing medium was re- moved and replaced with fresh culture medium.

At different time intervals, cells were analyzed for cell cycle progression. Data show that treat- ment with paclitaxel caused G2/M arrest at approximately 4 h posttreatment, maximizing at 24 h. This was followed by a timely reversal of G2/M arrest to the baseline level by 48 h. b Cells were treated with paclitaxel for 3 h using pulsed dose schedules: Drug treatment with 0.33 µM on day 1 (B3); or day 1 and day 3 (B4);

or day 1, day 3, and day 5 (B5). Drug-containing culture medium was removed after treatment and replaced with fresh maintenance medium.

The cells were analyzed for cell cycle distribu- tion at 24 h after drug removal. B1 shows the cell cycle distribution of baseline without drug treatment. B2 shows the maximal arrest of G2/

M 24 hours after treatment with 1.0 µM pacli- taxel. B3–B5 show that treatment with pulsed paclitaxel three times a week using one-third of the initial dose sustained the G2/M cell cycle effect. Adapted from Chen et al. (2003a), and used with permission

a

b

(5)

tive or synergistic mechanism. Even in the subad- ditive situation, delaying radiation resulted in fewer surviving clones than immediate irradiation after drug treatment.

11.6.2.6

Clinical Protocol of Pulsed Paclitaxel and Radiation

A phase I/II clinical study for stage III NSCLC was designed taking into account the pharmacokinetic information, the timing of radiosensitization, and the cell cycle and apoptotic effect of paclitaxel from the preclinical studies. A 1 h infusion time was chosen to allow for a short exposure time to the drug for sensitization, followed by a rapid clearing of plasma drug concentration to avoid sensitizing normal tissues and to allow for less cytotoxicity to bone marrow. The clinical trial design dictated low-dose paclitaxel infusion in the morning on Monday, Wednesday, and Friday. Thoracic radia- tion (XRT) was given after 4:00 PM on the days when patients received paclitaxel, to allow for a minimum of a 4 h interval for cell cycle progres- sion. On Tuesday and Thursday, when there was no paclitaxel treatment, XRT was given any time after 11:00 AM. For the phase I study, the starting dose of paclitaxel was 15 mg/m2 with a dose es-

calation in 5 mg/m2 increments. The average XRT dose was 60–65 Gy to gross disease and 45–58 Gy to microscopic disease, given at 1.8 Gy daily frac- tions over 6–7.5 weeks. Forty-one patients were enrolled (23 for the phase I study and 18 for the phase II study), and 33 completed treatment. Eight patients did not complete protocol treatments due to acute allergic reactions in three, distant disease spread during therapy in three, and two intercur- rent deaths unrelated to therapy. Stage distribution of patients with NSCLC was two in stage I, one in stage IIB, 15 in stage IIIA, and 22 in stage IIIB. One patient had stage II mesothelioma.

11.6.2.7

Tumor Response, In-Field Tumor Control, and Survival

Almost all tumors had a remarkable and durable re- sponse to therapy using this regimen. Table 11.6.1 demonstrates the tumor response in the phase I study.

Mean tumor shrinkage was 82%±14%, 84%±16%, and 84%±27% for dose levels I, II, and III, respec- tively, with an average primary tumor shrinkage at 4–6 weeks posttherapy of 83%±7% (95% confi dence interval). The overall locoregional tumor response rate in the phase I study was 100% (2/17 [12%] com- plete response and 15/17 [88%] partial response).

Fig. 11.6.4 demonstrates an example of radiographic tumor shrinkage 4 weeks after treatment in a patient with stage III NSCLC. Local control for those patients who completed radiotherapy in both phase I and II was 98%, with a median follow-up of 11 months. For patients who did not complete the 7.5-week protocol treatment, the survival was dismal. For those who completed the protocol treatment, the survival es- timate was 52% at 1 year, 40% at 2 years, and 21%

at 3 years, and survival for all patients enrolled was 46% at 1 year, 33% at 2 years, and 18 % at 3 years (Table 11.6.2). The in-fi eld local control was durable up to the last day of follow-up for most patients, which has been demonstrated by the PET scans ob- tained 3 years later in some patients (Chen et al.

2003a).

11.6.2.8 Toxicity

As anticipated, low-dose pulsed paclitaxel chemo- radiation was associated with low toxicity. There was no treatment interruption from side effects of

Fig. 11.6.3 Human cancer cell lines A431, A549, and NCI-H520 were treated with paclitaxel at 200 nM, 600 nM, or 2 µM con- centrations. Cells were analyzed for apoptosis using Tunnel assay at 24 h as well as 48 h after drug treatment. Data showed that apoptosis was more prominent at a higher drug concentra- tion and was observed primarily at 48 h after drug treatment.

Adapted from Chen et al. (2003a), and used with permission

(6)

therapy. Toxicity was assessed for all patients in the phase I study, including one patient who re- ceived only two-thirds of the total radiation dose.

There were no patients who experienced grade 3 or 4 neutropenia, thrombocytopenia, neuropathy, or cardiac arrhythmia. Three of 18 patients (17%) experienced grade 3 pneumonitis, and 3/18 (17%) experienced grade 3 esophagitis. No patients expe- rienced grade 4 pneumonitis or esophagitis (Chen et al. 2001).

11.6.2.9

Future Directions in Chemopotentiation of Radiotherapy for Stage III NSCLC

At least eight different dosing schedules for pacli- taxel chemoradiation have been employed in clinical trials for treating stage III NSCLC; as a whole, they

Table 11.6.1 Tumor response 4–6 weeks after pulsed low-dose paclitaxel chemoradiation. Adapted from Chen et al. (2003a) with permission. (CR complete response [disappearance of tumor], PR partial response [>50% volume reduction])

Pulsed paclitaxel Tumor Response rate dose level shrinkage

CR PR

I, 15 mg/m2 (n=6) 82%±14% 2 4 II, 20 mg/m2 (n=6) 84%±16% 0 6 III, 25 mg/m2 (n=4) 84%±27% 0 4

Average 83%±7% (95% CI) 14% 86%

Fig. 11.6.4 Tumor response by CT scans. A large lesion of non-small cell lung cancer presented with superior vena cava compression in the right upper lobe of the lung. The lesion had completely disappeared 4 weeks after pulsed paclitaxel chemoradiation. Adapted from Chen et al. (2003), and used with permission

Table 11.6.2 Survival and local tumor control comparisons for stage III NSCLC treated with combination chemoradia- tion. Adapted from Chen et al. (2003a) with permission (RT radiotherapy)

Chemoradiation trials 2-year 3-year Local

survival survival control

Chen et al.: Pulsed Taxol and RT

All patients 33% 18% N/A

Patients completing therapy 40% 21% 98%

Schaake-Koning et al. (EORTC)

Chemoradiation arm 26% 16% 30%a

RT arm 13% --- 19%

Furuse et al. (Japanese)

Concurrent chemo arm 35% 22% --- Sequential chemo arm 27% 15% ---

Dillman et al. (CALGB)

Chemoradiation arm 26% 23% ---

RT arm 13% 11% ---

Arriagada et al. (French)

Chemoradiation arm 21% 11% 17%b

RT arm 14% 5% 15%

Sause et al. (RTOG8808)

Chemoradiation arm 32% 17% ---

RT arm 22% 11% ---

Morton et al. (NCCT)

Chemoradiation arm 21% --- 46%c

RT arm 16% --- 45%

a Actuarial local control at 2 years

b Local control at 1 year

c Local control at time of fi rst relapse

pre-treatment

1 month post-treatment

(7)

targeted therapy, gene therapy, hypoxic cell-targeted therapy, or other innovative therapeutic approaches may help prevent distant metastasis.

11.6.3

Cytokine Markers for Radiation Pulmonary Injury

11.6.3.1

Radiation Lung Injury

Due to normal tissue constraints, high-dose radia- tion to the tumoricidal dose for chest tumors has not been feasible using standard external beam ra- diation therapy. For standard radiation treatment to the chest, radiation dose has been limited to 60–65 Gy in order to avoid serious pneumonitis, esophageal stricture, fi stula, or cardiac damage. Indeed, normal tissue injury from combined modality treatment for lung cancer has been the major dose-limiting factor for aggressive chemoradiation treatment. Through clinical experience using chemoradiation combined modality therapy for lung cancer, higher rates of both esophagitis and pneumonitis have been consistently observed (Chen and Okunieff 2004). Although interstitial pneumonitis has been recognized as a distinct clinical complication of cancer therapy, no routine diagnostic testing has been established as a simple way to assess the risk of pneumonitis prior to cancer treatment. Although most interstitial pneu- monitis from cancer therapy is self-limiting, seri- ous and potentially lethal incidences have been ob- served following all treatment modalities, including radiation therapy (Yorke et al. 2002; Jenkins et al.

2003; Ash-Bernal et al. 2002; Reckzeh et al. 1996), chemotherapy (Thomas et al. 2000; Sleijfer 2001;

Wang et al. 2001), and even some of the more recently developed molecularly-targeted therapies (Rosado et al. 2003; Burton et al. 2003).

Among all types of interstitial pneumonitis caused by different agents, radiation-induced pneumonitis has been the most widely investigated, both in clini- cal and in laboratory research. The peak incidence of radiation pneumonitis is between 6 weeks and 3 months after completion of radiation treatments.

However, it can also occur unexpectedly, with little or no warning; therefore, many attempts have been made to identify clinical risk factors for its onset. Clinical studies have reported a number of clinical factors, including total radiation dose, irradiated lung vol- ume exceeding 20 Gy, mean lung dose, fractionation, have shown both mixed response rates and toxic-

ity profi les. These schedules include concurrent ra- diation with continuous infusion of paclitaxel, daily paclitaxel, twice-weekly paclitaxel, thrice-weekly pa- clitaxel, weekly paclitaxel, and once-every-3–4-weeks paclitaxel as a single agent or in combination with a second chemotherapeutic agent. The overall response rates have been in the range of 65–100%, but gener- ally treatment toxicities also have been high, particu- larly in neutropenia, thrombocytopenia, esophagitis, and pneumonitis (Chen and Okunieff 2004). These effects are attributed to the radiosensitizing effect of paclitaxel on normal tissues as well.

Stage III NSCLC often presents with large tumor volume for the primary and the regional nodal dis- ease. If the primary and regional tumors are not con- trolled, distant metastasis cannot be prevented. The lack of effective local therapy for inoperable NSCLC has been well recognized. Biopsy of tumors after ther- apy for locally advanced NSCLC resulted in only 17%

local tumor control after chemoradiation therapy and only 15% after radiotherapy alone (Arriagada et al. 1991). Although chemoradiation is the current standard of care for stage III NSCLC, improvements in local disease control and survival have been mod- est, despite aggressive clinical investigations using numerous chemotherapeutic agents in conjunction with radiation in many phase I/II studies (Chen and Okunieff 2004). Among these dose schedules, the pulsed paclitaxel chemoradiation schedule, which was based on the preclinical research model, yielded superior primary tumor response and ultimate local control. These results support the idea that preclini- cal studies using cancer cell lines or animal models to address the sequence and timing of treatment modalities may prove more fruitful in gaining favor- able clinical outcomes than those regimens built on an empirical basis. The superior local tumor control produced by pulsed paclitaxel chemoradiation for stage III NSCLC has provided new information in the understanding of cancer biology for this tumor stage. This regimen has overcome the local failure of stage III NSCLC for primary chest tumors as large as 10–12 cm at the time of presentation. The differential between the signifi cant gain of local disease control (98% by pulsed paclitaxel vs. historical control of large randomized studies of 46% at best) vs. the modest survival gain (3-year survival of 21% vs. 15% of large studies of historical control) emphasizes that future therapy should aim not only for chemopotentiation for chest disease control, but also for more effective prevention and treatment of distant micrometastasis (Table 11.6.2). Antiangiogenic therapy, molecularly

(8)

daily fraction size, performance status, pretreatment pulmonary function, gender, low pretreatment blood oxygen, high C-reactive protein, and others (Roach et al. 1995; Kwa et al. 1998; Graham et al. 1999;

Inoue et al. 2001; Segawa et al. 1997; Robnett et al.

2000; Hernando et al. 2001). However, despite the identifi cation of these clinical contributing factors, clinical research has not led to the development of a reliable and simple diagnostic laboratory test that could predict the risk of postradiation pneumonitis and, in particular, one that could be administered be- fore starting therapy. The development of such a test using patient blood specimens or lavage fl uid from bronchial washing is highly desirable because of the unpredictable nature of serious adverse events, which occur sporadically and without reliable clinical warn- ing signs; its use prior to or during the early phase of therapy would allow clinicians to customize and modify therapy intensity for those patients at higher risk for serious interstitial pneumonitis.

11.6.3.2

Cytokine and Radiation Lung Injury

Progress made in recent years has increased our un- derstanding of the cellular and molecular mecha- nisms involved in radiation lung injury. We have come to appreciate the complexity of radiation pneumoni- tis, now seen as a process involving active interaction among resident cells of the lung parenchyma and circulatory immune and infl ammatory cells. There is increasing evidence of immune cells augmenting pneumonitis through complex autocrine, paracrine, and systemic regulatory mechanisms critically or- chestrated by cytokines. Early studies suggested that radiation-induced lung injury was characterized by alveolar infi ltrates of mononuclear cells, primarily CD4+ T cells and macrophages/monocytes (mono- nuclear alveolitis), while exhibiting a relative lack of neutrophils (Fryer et al. 1978; Roberts et al. 1993), a common marker for infectious processes. In addition, several studies using bronchoscopy have confi rmed this fi nding in patients with active pneumonitis, dem- onstrating the presence of mononuclear cells with- out signifi cant numbers of neutrophils in the lavage fl uids (Maasilta et al. 1993; Nakayama et al. 1996).

Ultimately, mononuclear alveolitis is followed by pro- gressive fi brosis of the lung as well as the deposition and accumulation of collagen fi bers and extracellular matrix. Clinically, a decline in lung volume, compli- ance, and diffusion capacity is an unavoidable long- term consequence of lung fi brosis.

Animal research has induced multiple humoral factors (cytokines) in the lung by ionizing radiation (Rubin et al. 1995; Johnston et al. 1998; Hallahan et al. 1997, 2002). Rubin et al. (1995) reported a cas- cade of cytokine induction including interleukin 1α (IL-1α), transforming growth factor ß1 (TGF-ß1), and TGF-ß2 in lung tissue after radiation in animal research models. Because most animal research has been conducted in large single fractions of radiation, the relevance to cancer patients receiving fractionated daily low-dose radiation remains unclear. However, based on data from animal research models, it is hy- pothesized that when normal lung tissue is exposed to chemotherapy, radiation, oxygen, tumor necrosis factor (TNF-α), or other foreign insult such as lipo- polysaccharide, a cascade of cellular and humoral events occurs as a tissue response to injury (Chen et al. 2003). There is interaction among alveolar epi- thelium (type I and type II pneumocytes), vascular endothelium, fi broblasts, lymphocytes, and macro- phages through the humoral factors such as adhe- sion molecules, chemokines, infl ammatory cytokines (TNF-α, IL-6, and IL-1), and fi brotic cytokines (basic fi broblast growth factor [bFGF], TGF-ß, and plate- let-derived growth factor [PDGF]). Chemokines and adhesion molecules mediate leukocyte traffi cking, extravasation from the vascular compartment, and homing to sites of infl ammation (Kaseda et al. 2000;

Butcher et al. 1996; Hallahan et al. 1997; Ding et al. 2000). Macrophages and lymphocytes are further recruited from the bone marrow to the chest, causing alveolar infi ltrates during the pneumonitic phase.

At least three different classes of cytokines have been reported to correlate with the risk of radiation pneumonitis in patients: the infl ammatory cytokines IL-1α and IL-6, the profi brotic cytokine TGF-β1, and intercellular adhesion molecule-1 (ICAM-1). These cytokines will be discussed in further detail.

11.6.3.3

Infl ammatory Cytokines IL-1α and IL-6

Radiation pneumonitis can be regarded as an ab- errant infl ammatory response to radiation injuries.

Both IL-1α and IL-6 are cytokines central to regula- tion of immunity and infl ammatory responses. These cytokines are important immunoregulatory moieties and share some common immune functions. Both cytokines are pleiotropic infl ammatory cytokines, recognized as "acute phase response" proteins. They are chemotactic for mononuclear cells, and they ac- tivate lymphocytes, regulate fevers, and precipitate a

(9)

fi brovascular response. While the source of IL-1α is primarily from monocytes and alveolar macrophages (Elias et al. 1985; O’Brien-Lander et al. 1993), IL- 6 is synthesized by a variety of cells, including the alveolar macrophages, endothelium, type II pneumo- cytes, T lymphocytes, and lung fi broblasts (Kelley 1990; Cromwell et al. 1992; Crestani et al. 1994).

In vitro experiments have shown that alveolar macrophages exposed to radiation release increased quantities of IL-1α and IL-1β (O’Brien-Lander et al. 1993). It has also been shown that IL-1α stimu- lates human lung fi broblasts to produce IL-6 and sta- bilizes IL-6 messenger RNA production (Elias and Lentz 1990). Others have reported IL-1 induction of IL-6 in a variety of cells, including thymocytes, and have suggested that IL-6 is involved in many of the pleiotropic effects of IL-1 (Helle et al. 1988). Such in vitro work indeed suggests a regulatory relation- ship between the two infl ammatory cytokines. Chen et al. (2001a, 2002) analyzed a panel of cytokines in serial blood samples of cancer patients receiving thoracic radiation. Radiation pneumonitis was diag- nosed using the National Cancer Institute common toxicity criteria. Cytokine analysis was assayed for IL-1α, IL-6, MCP-1, E-selectin, L-selectin, P-selectin, TGF-ß1, and basic bFGF, using enzyme linked immu- nosorbent assay (ELISA). Among all these cytokines analyzed, only the two infl ammatory cytokines, IL-6 and IL-1α, were persistently higher in patients who subsequently developed pneumonitis after radiation.

Additionally, patients with high pretreatment circu- lating IL-6 and IL-1α levels had a greater chance of developing subsequent pneumonitis after therapy (Fig. 11.6.5), suggesting the possibility of using pre- treatment IL-1α and IL-6 levels to predict patients at risk for radiation pneumonitis.

11.6.3.4

Chemokines and Cell Adhesion Molecules

Tissue infl ammation requires leukocytes to undergo transendothelial migration and extravasation of the vascular compartment into sites of infl ammation.

Chemokines and adhesion molecules are key cyto- kines in facilitating leukocyte recruitment to the site of infl ammation (Kaseda et al. 2000; Butcher et al. 1996; Hallahan et al. 1997; Ding et al. 2000).

Johnston et al. (1998) investigated a panel of che- mokines including monocyte chemotactic protein (MCP)-1, lymphotactin (Ltn), RANTES, eotaxin, macrophage infl ammatory protein (MIP-1α, -1ß, and -2), and interferon-inducible protein 10 (IP-10)

in fi brosis-prone mice (C57BL/6) and fi brosis-resis- tant mice (C3H/HeJ). In these studies, local MCP-1 and IP-10 expressions did not increase 8 weeks af- ter lung radiation, but a signifi cantly higher level of expression was found only in the fi brosis-prone mice 26 weeks after lung radiation. Hallahan et al.

(1997) investigated the role of adhesion molecules in animal lungs after radiation. They found an increase of ICAM-1 and E-selectin expression in vascular endothelium after radiation. In the animal model, they found that anti-ICAM-1 blocking antibody at- tenuated the infl ammatory cell infi ltration to the lungs of mice after radiation exposure. In addition, using ICAM-1 knockout mice, which were defi cient in expressing ICAM-1 after radiation, radiation-in- duced pulmonary infl ammatory cell infi ltration was abrogated (Hallahan et al. 1997a, 2002). Research in chemokine and adhesion molecules in cancer patients produced some correlated fi ndings. Ishii and Kitamura (1999) reported that patients with an increase of ICAM-1 in circulating cytokine lev- els during radiation had a higher risk of radiation

Fig. 11.6.5 a Circulating IL-1α (in pg/ml) before radiation, weekly during radiation, and after completing radiation treat- ments. b Circulating IL-6 (in pg/ml) before radiation, weekly during radiation, and after completing radiation treatments.

Adapted from Chen et al. (2001a) and from Chen et al. (2002);

used with permission from both publishers

b a

(10)

pneumonitis. Chen et al. (2002) investigated the role of other chemokine and adhesion molecules in the blood of cancer patients, including MCP-1, E-selectin, L-selectin, and P-selectin, but they found no correla- tion with the risk of radiation lung injury.

11.6.3.5

Fibrotic Cytokine: Transforming Growth Factor β TGF-ß is a key modulator in the induction of fi brosis, alterations in collagen metabolism, and increase in extracellular matrix formation following radiation therapy (Burger et al. 1998; Barcelloss-Hoff 1998; Randall and Coggle 1995; Rodemann and Bamberg 1995). TGF-ß has been shown to be a growth factor regulating fi broblast proliferation, differentiation, and matrix production under physi- ological as well as pathophysiological conditions (Kovacs 1991; Fine and Goldstein 1987; Sigel et al. 1996; Zhang and Jacobberger 1996). It was found to be a primary cytokine for radiation-induced fi brosis (Rodemann and Bamberg 1995). In animal research models, different investigators have found that TGF-ß expression is induced after lung radia- tion and is the major mediator for postradiation lung fi brosis (Finkelstein et al. 1994; Rubin et al. 1992;

Franko et al. 1997). The search for TGF-ß as a marker for radiation lung injury in patients has been com- plicated by the fact that lung cancers may produce TGF-β. Nonetheless, Anscher et al. (1998) found a correlation between higher risk of radiation lung in- jury in patients with high circulating levels of TGF-β near the end of radiotherapy. This observation was signifi cant for the patients with normal pretreatment TGF-β levels but not signifi cant for those with higher than normal pretreatment TGF-β levels.

11.6.4 Conclusions

For decades, radiobiologists and radiation oncolo- gists have searched for ways to enhance radiation effects on human solid tumors. Fletcher (1973) re- ported the clinical dose-response curves of human malignant epithelial tumors based on clinical expe- riences at M.D. Anderson Cancer Center in Houston.

In this report, the volume effect on tumor control probability was explored, and the concept of larger tumors requiring higher radiation doses to effec- tively control the tumor by radiation was introduced.

Oncologists at the University of Florida also reported a similar fi nding for squamous cell carcinoma of the head and neck region. The dose required to eradicate solid tumors is proportional to the tumor volume:

60–65 Gy for tumors smaller than 1.0 cm in diam- eter, 65–70 Gy for tumors 1.5–2.0 cm, 70–75 Gy for tu- mors 2.5–3.0 cm, and 75–80 Gy for tumors 3.5–6.0 cm (Million et al. 1994). If one extrapolates Fletcher’s estimate, it would require an 80–100 Gy radiation dose to eradicate large epithelial tumors of stage III NSCLC. Such a high dose to the chest has not been possible in the clinical setting due to potential fatal pneumonitis. Clinical practice in treating stage III NSCLC in general has kept radiation doses below 65 Gy and has therefore produced unsatisfactory lo- cal regional disease control for patients in this stage.

If gross tumors in the chest cannot be eradicated, distant failures are defi nite sequelae.

Thus, optimization of chemotherapy and radiation continues to deserve preclinical studies aimed at maximizing the primary tumor control while keeping the toxicity low. For enhancing local tumor control, more frequent but lower doses of drug appear par- ticularly promising in maximizing tumor response and minimizing toxicity. An interesting observation was made in this phase I study of pulsed paclitaxel chemoradiation: The lack of dose response with in- creasing paclitaxel doses beyond 15 mg/m2 supports that when low-dose chemotherapy is used as a ra- diation-sensitizing agent, escalating chemotherapy doses may not further potentiate radiation but rather may increase the toxicity of therapy. The concept of a

“minimally effective dose” for radiation potentiation by chemotherapy is contradictory to the traditional strategy of systemic chemotherapy when achieving a maximally tolerated dose is the primary goal. The su- perior tumor response and the durable tumor control achieved by pulsed low dose paclitaxel chemoradia- tion are intriguing. This dramatic local tumor effect cannot be explained by cell cycle and apoptotic ef- fects of pulsed paclitaxel alone. More recently, small and frequent dosing of chemotherapeutic agents has been shown to suppress tumor growth through the mechanism of antiangiogenesis in animal tumor models (Boehm et al. 1997; Browder et al. 2000).

Other potential mechanisms that may contribute to the effectiveness of the small frequent dosing sched- ule of paclitaxel and radiation should be further in- vestigated in animals.

Recent discoveries in translational research using specimens from lung cancer patients have allowed a major breakthrough in our understanding of the so- matic mutation of epidermal growth factor receptor

(11)

(EGFR) in relation to tumor response to treatments by gefi tinib (Iressa). Clinically, gefi tinib offers an overall 10% response rate for patients with NSCLC.

Examinations of the EGFR genes have revealed that mutations at the tyrosine kinase domain not only spe- cifi cally predicted responders to gefi tinib treatments but also showed a striking correlation with patient characteristics: Mutations were more frequent in ad- enocarcinomas than in other NSCLC, more frequent in women than in men, and more frequent in patients from Japan than in those from the U.S. (Lynch et al.

2004; Paez et al. 2004). Serious lung injury was infre- quently observed in large studies of patients treated with gefi tinib for NSCLC, and included rare but fatal interstitial pneumonitis occurring with higher inci- dence in the Japanese patients (1.87%) than in pa- tients outside Japan (0.35%).

Such fi ndings support endeavors in identifying gene and molecular markers, not only for the effi - cacy of therapy but also for identifying markers for normal tissue injury. Simultaneous measurement of normal tissue consequences in translational research is critical, as the ultimate goal of treatment must in- clude minimizing the acute and long-term toxicity to normal tissues. Cancer therapy has evolved to- wards a multiagent and multitarget approach, with the inclusion of gene therapy, antiangiogenic therapy, inhibition of epidermal growth factor pathways, in- hibition of oncogene pathways, inhibition of other signal transduction pathways, hypoxic cell target- ing therapy, and others (Chen et al. 2003). Balancing therapeutic effects versus normal tissue effects will become more complex and more challenging to on- cologists in the future.

References

Anscher MS, Kong F-M, Andrews K, et al. (1998) Plasma trans- forming growth factor ß1 as a predictor of radiation pneu- monitis. Int J Radiat Oncol Biol Phys 41:1029–1035 Arriagada R, le Chevalier T, Quoix E, et al. (1991) ASTRO

Plenary: effect of chemotherapy on locally advanced non- small cell lung carcinoma: a randomized study of 353 patients. Int J Radiat Oncol Biol Phys 20:1183–1190 Ash-Bernal R, Browner I, Erlich R (2002) Early detection and

successful treatment of drug-induced pneumonitis with corticosteroids. Cancer Invest 20:876–879

Barcelloss-Hoff M-H (1998) How do tissues respond to damage at the cellular level? The role of cytokines in irradiated tis- sues. Radiat Res 150:109–120

Boehm T, Folkman J, Browder T, et al. (1997) Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature 390:404–407

Brodin O, Wagenius G, Helsing M (2000) Daily paclitaxel (P) and radiotherapy (RT) in patients (PTS) with locally

advanced non-small cell lung cancer (NSCLC). A phase I study. [abstract 2022]. In: Program/proceedings of the American Society of Clinical Oncology, 36th annual meet- ing, New Orleans. ASCO, Virginia, p 516a

Browder T, Butterfi eld CE, Kraling BM, et al. (2000) Antian- giogenic scheduling of chemotherapy improves effi cacy against experimental drug-resistant cancer. Cancer Res 60:1878–1886

Burger A, Löffl er H, Bamberg M, et al. (1998) Molecular and cel- lular basis of radiation fi borsis. Int J Radiat Biol 73:401–408 Burton C, Kaczmarski R, Jan-Mohamed R (2003) Interstitial

pneumonitis related to rituximab therapy. N Engl J Med 348:2690–2691

Butcher EC, Picker LJ (1996) Lymphocyte homing and homeo- stasis. Science 272:60–66

Chen Y, Okunieff P (2004) Radiation and third generation che- motherapy. Hematol Oncol Clin N Am 18:55–80

Chen Y, Okunieff P, Ahrendt SA (2003) Translational research in lung cancer. Semin Surg Oncol 21:205–219

Chen Y, Pandya K, Keng PC, et al. (2001) Schedule dependent pulsed low-dose paclitaxel radiosensitization for thoracic malignancy. Am J Clin Oncol 24:432–437

Chen Y, Pandya K, Keng PC, et al. (2003a) Phase I/II clinical trial using pulsed low-dose paclitaxel radiosensitization for thoracic malignancies: a therapeutic approach based on pre-clinical research of human lung cancer cells. Clin Cancer Res 9:969–975

Chen Y, Rubin P, Williams J, et al. (2001a) Circulating IL-6 as a predictor of radiation pneumonitis. Int J Radiat Oncol Biol Phys 49:641–648

Chen Y, Williams J, Ding I, et al. (2002) Radiation pneumoni- tis and early circulatory cytokine markers. Semin Radiat Oncol 12:26–33

Choy H, Akerly W, Safran H, et al. (1994) Phase I trial of out- patient weekly paclitaxel and concurrent radiation ther- apy for advanced non-small cell lung cancer. J Clin Oncol 12:2682–2686

Choy H, Rodriguez FF, Koester S, et al. (1993) Investigation of taxol as a potential radiation sensitizer. Cancer 71:3774–3778 Cromwell O, Hamid O, Corrigan CJ, et al. (1992) Expression

and generation of interleukin-8, IL-6, and granulocyte macrophage colony-stimulating factor by bronchial epithe- lial cells and enhancement by IL-1β and tumor necrosis factor α. Immunology 77:330–337

Crestani B, Cornillet P, Dehoux M, et al. (1994) Alveolar type II epithelial cells produce interleukin-6 in vitro and in vivo. J Clin Invest 94:731–740

Curran WJ Jr, Scott C, Langer C, et al. (2000) Phase III com- parison of sequential vs. concurrent chemoradiation for PTS with unresected stage III non-small cell lung cancer (NSCLC): initial report of Radiation Therapy Oncology Group (RTOG) 9410. Proc ASCO 19:484a abstr 1891 Dillman RO, Seagren SL, Propert KJ, et al. (1990) A random-

ized trial of induction chemotherapy plus high dose radia- tion versus radiation alone in stage III non-small cell lung cancer. N Engl J Med 323:940–945

Ding Z, Xiong K, Issekutz TB (2000) Regulation of chemokine- induced transendothelial migration of T lymphocytes by endothelial activation: differential effects on naïve and memory T cells. J Leukoc Biol 67:825–833

Elias JA, Lentz V (1990) IL-1 and tumor necrosis factor syner- gistically stimulate fi broblast IL-6 production and stabilize IL-6 messenger RNA. J Immunol 145:161–166

(12)

Elias JA, Schreiber AD, Gustilo K, et al. (1985) Differential interleukin-1 elaboration by unfractionated and density fractionated human alveolar macrophages and blood monocytes: relationship to cell maturity. J Immunol 135:3198–3204

Elomaa L, Joensuu H, Kulmala J, et al. (1995) Squamous cell carcinoma is highly sensitive to taxol, a possible new radia- tion sensitizer. Acta Otolaryngol 115:340–344

Fine A, Goldstein RH (1987) The effect of transforming growth factor beta on cell proliferation and collagen formation by lung fi broblasts. J Biol Chem 262:3897–3902

Finkelstein JN, Johnston CJ, Baggs R, et al. (1994) Early altera- tions in extracellular matrix and transforming growth factor ß gene expression in mouse lung indicative of late radiation fi brosis. Int J Radiat Oncol Biol Phys 28:621–631 Fletcher GH (1973) Clinical dose-response curves of human

malignant epithelial tumours. Br J Radiol 46:1–12 Franko AJ, Sharplin J, Ghahary A, et al. (1997) Immunihisto-

chemical localization of transforming growth factor beta and tumor necrosis alpha in the lungs of fi brosis-prone and

“non-fi brosing” mice during the latent period and early phase after irradiation. Radiat Res 147:245–256

Fryer CJH, Fitzpatrick PJ, Rider WD, et al. (1978) Radiation pneumonitis: experience following a large single dose of radiation. Int J Radiat Oncol Biol Phys 4:931–936

Furuse K, Fukuoka M, Kawahara M, et al. (1999) Phase III study of concurrent versus sequential thoracic radiotherapy in combination with mitomycin, vindesine, and cisplatin in unresectable stage III non-small-cell lung cancer. J Clin Oncol 17:2692–2699

Geard CR, Jones JM, Schiff PB (1993) Taxol and radiation. J Natl Cancer Inst Monographs 15:89–94

Gianni L, Kearns CM, Giani A, et al. (1995) Nonlinear phar- macokinetics and metqabolism of paclitaxel and its phar- macokinetic/pharmacodynamic relationships in humans. J Clin Oncol 13:180–190

Graham MV, Burdy JA, Emami B, et al. (1999) Clinical dose- volume histogram analysis for pneumonitis after 3D treat- ment for non-small cell lung cancer. Int J Radiat Oncol Biol Phys 45:323–329

Grant D, Williams TL, Zahaczewsky M, et al. (2003) Compari- son of antiangiogenic activities using paclitaxel (Taxol) and docetaxel (Taxotere). Int J Cancer 104:121–129 Hallahan DE, Geng L, Shyr Y (2002) Effects of intercellular

adhesion molecule 1 (ICAM-1) null mutation on radiation- induced pulmonary fi brosis and respiratory insuffi ciency in mice. J Natl Cancer Inst 94:733–741

Hallahan DE, Virudachalam S (1997) Ionizing radiation medi- ates expression of cell adhesion molecules in distinct histo- logical patterns within the lung. Cancer Res 57:2096–2099 Hallahan DE, Virudachalam S (1997a) Intercellular adhesion

molecule-1 knockout abrogates radiation induced pulmo- nary infl ammation. Proc Natl Acad Sci USA 94:6432–6437 Havemann K, Wolf M, George C, et al. (1995) Paclitaxel and

simultaneous radiation in the treatment of stage III A/B non-small cell lung cancer. Semin Oncol 22:19–22 Helle M, Brakenhoff JPJ, De Groot ER, et al. (1988) Interleu-

kin 6 is involved in interleukin 1-induced activities. Eur J Immunol 18:957–959

Hernando ML, Marks LB, Bentel GC, et al. (2001) Radiation- induced pulmonary toxicity: a dose-volume histogram analysis in 201 patients with lung cancer. Int J Radiat Oncol Biol Phys 51:650–659

Hornback NB, Shen R-N, Sutton GP, et al. (1994) Synergistic cytotoxic and antitumor effects of irradiation and Taxol on human Hela cervix carcinoma and mouse B16 melanoma cells. In Vivo 8:819–824

Inoue A, Kunitoh H, Sekine I, et al. (2001) Radiation pneumo- nitis in lung cancer patients: a retrospective study of risk factors and the long-term prognosis. Int J Radiat Oncol Biol Phys 49:649–655

Ishii Y, Kitamura S (1999) Soluble intercellular adhesion mol- ecule-1 as an early detection marker for radiation pneumo- nitis. Eur Respir J 13:733–738

Jenkins P, D’Amico K, Benstead K, et al. (2003) Radiation pneu- monitis following treatment of non-small-cell lung cancer with continuous hyperfractionated accelerated radiother- apy (CHART). Int J Radiat Oncol Biol Phys 56:360–366 Johnston CJ, Wright TW, Rubin P, et al. (1998) Alterations in the

expression of chemokine mRNA levels in fi brosis-resistant and -sensitive mice after thoracic irradiation. Exp Lung Res 24:321–337

Jordan MA, Wendell K, Gardiner S, et al. (1996) Mitotic block induced in HeLa cells by low concentrations of paclitaxel results in abnormal mitotic exit and apoptotic cell death.

Cancer Res 56:816–825

Kaseda M, Kadota J, Mukae H, et al. (2000) Possible role of L- selectin in T lymphocyte alveolitis in patients with active pulmonary sarcoidosis. Clin Exp Immunol 121:146–150 Kelley J (1990) Cytokines of the lung. Am Rev Respir Dis

141:765–788

Keng PC, Okunieff P, Chen Y (2000) Low dose taxol/taxotere radiosensitization for human lung cancer cells is schedule dependent. Proceedings of the 42nd annual ASTRO meet- ing. Int J Radiat Oncol Biol Phys 48:272

Kirkbride P, Gelmon K, Eisenhauer E (1999) Paclitaxel and concurrent radiotherapy in locally advanced non-small cell lung cancer: the Canadian experience. Semin Radiat Oncol 9(suppl 1):102–107

Kovacs EJ (1991) Fibrogenic cytokines: the role of immune mediators in the development of scar tissue. Immunol Today 12:17–23

Kumar N (1981) Taxol-induced polymerization of purifi ed tubu- lin. Mechanism of action. J Biol Chem 256:10435–10441 Kwa SLS, Lebesque JV, Theuws JCM, et al. (1998) Radiation pneu-

monitis as a function of mean lung dose: an analysis of pooled data of 540 patients. Int J Radiat Oncol Biol Phys 42:1–9 Lau DH, Ryu JK, Gandara DR, et al. (1997) Twice weekly pacli-

taxel and radiation for stage III non-small-cell lung cancer.

Semin Oncol 24(suppl 12):106–109

Lokeshwar BL, Ferrell SM, Block NL (1995) Enhancement of radiation response of prostatic carcinoma by taxol: thera- peutic potential for late-stage malignancy. Anticancer Res 15:93–98

Lynch T, Bell D, Sordella R, et al. (2004) Activating mutations in the epidermal growth factor receptor underlying respon- siveness of non-small cell lung cancer to gemfi tinib. N Engl J Med 350:2129–2139

Maasilta P, Hallman M, Taskinen E, et al. (1993) Bronchoal- veolar lavage fl uid fi ndings following radiotherapy for non-small cell lung cancer. Int J Radiat Oncol Biol Phys 26:117–123

Manfredi JJ, Parness J, Horwitz SB (1982) Taxol binds to cel- lular microtubules. J Cell Biol 94:688–696

Milas L, Hunter NR, Kurdoglu B, et al. (1995) Kinetics of mitotic arrest and apoptosis in murine mammary and

(13)

ovarian tumors treated with taxol. Cancer Chemother Pharmacol 35:297–303

Milas L, Hunter NR, Mason KA, et al. (1995a) Tumor re-oxy- genation as a mechanism of taxol-induced enhancement of tumor radioresponse. Acta Oncologica 34:409–412 Million RR, Cassisi NJ, Mancuso AA, et al (1994). Chapter 6:

Management of the neck for squamous cell carcinoma. In:

Million RR, Cassisi NJ (eds) Management of head and neck cancer, a multidisciplinary approach, 2nd edn. Lippincott, Philadelphia, pp 75–142

Milross CG, Mason KA, Hunter NR, et al. (1996) Relationship of mitotic arrest and apoptosis to antitumor effect of pacli- taxel. J Natl Cancer Inst 88:1308–1314

Morton RF, Jett JR, McGinnis WL, et al. (1991) Thoracic radia- tion therapy alone compared with combined chemother- apy for locally unresectable non-small cell lung cancer. Ann Int Med 115:681–686

Murphy WK, Fossella FV, Winn RJ, et al. (1993) Phase II study of Taxol in patients with untreated non-small cell lung cancer. J Natl Cancer Inst 85:384–388

Nakayama Y, Makino S, Fukuda Y, et al. (1996) Activation of lavage lymphocytes in lung injuries caused by radiotherapy for lung cancer. Int J Radiat Oncol Biol Phys 34:459–467 O’Brien-Lander A, Nelson ME, Kimler BF, et al. (1993) Release

of interleukin-1 by human alveolar macrophages after in vitro irradiation. Radiat Res 136:37–41

Parness J, Horwitz SB (1981) Taxol binds to polymerized tubu- lin in vitro. J Cell Biol 91:479–487

Paez JG, Janne PA, Lee JC, et al. (2004) EGFR mutations in lung cancer: correlation with clinical response to gefi tinib therapy. Science 304:1497–1500

Pukkinen JO, Elomaa L, Joensuu H, et al. (1996) Paclitaxel- induced apoptotic changes followed by time-lapse video microscopy in cell lines established from head and neck cancer. J Cancer Res Clin Oncol 122:214–218

Randall K, Coggle JE (1995) Expression of transforming growth factor ß1 in mouse skin during the acute phase of radiation damage. Int J Radiat Biol 68:301–309

Rathmann J, Leopold KA, Rigas JR (1999) Daily paclitaxel and thoracic radiation therapy for non-small-cell lung cancer:

preliminary results. Semin Radiat Oncol 9(suppl 1):130–135 Reckzeh B, Merte H, Pfl uger KH, et al. (1996) Severe lymphocy- topenia and interstitial pneumonia in patients treated with paclitaxel and simultaneous radiotherapy for non-small cell lung cancer. J Clin Oncol 14:1071–1076

Roach M, Gandara D, You H-S, et al. (1995) Radiation pneumo- nitis following combined modality therapy for lung cancer:

analysis of prognostic factors. J Clin Oncol 13:2606–2612 Roberts CM, Foulcher E, Zaunders JJ, et al. (1993) Radiation

pneumonitis: a possible lymphocyte-mediated hypersensi- tivity reaction. Ann Intern Med 118:696–700

Robnett TJ, Machtay M, Vines EF, et al. (2000) Factors pre- dicting severe radiation pneumonitis in patients receiv- ing defi nitive chemoradiation for lung cancer. Int J Radiat Oncol Biol Phys 48:89–94

Rodemann HP, Bamberg M (1995) Cellular basis of radiation induced fi brosis. Radiother Oncol 35:83–90

Rodriguez M, Sevin BU, Perras J, et al. (1995) Paclitaxel: a radiation sensitizer of human cervical cancer cells. Gyne- col Oncol 57:165–169

Rosado MF, Donna E, Ahn YS (2003) Challenging problems in advanced malignancy: case 3. Imatinib mesylate-induced interstitial pneumonitis. J Clin Oncol 21:3171–3173

Rosenthal DI, Algazy K, Dowell J, et al. (2000) Seven-week continuous infusion paclitaxel concurrent with radiation therapy for locally advanced non-small cell lung cancer:

a phase I study [abstract 2164]. In: Program/proceedings of the American Society of Clinical Oncology, 36th annual meeting, New Orleans. ASCO, Virginia, p 549a

Rowinsky EK, Donehower RC, Jones RJ, et al. (1988) Microtu- bule changes and cytotoxicity in leukemia cell lines treated with taxol. Cancer Res 48:4093–4100

Rubin P, Finkelstein J, Shapiro D (1992) Molecular biology mechanisms in the radiation induction of pulmonary injury syndromes: interrelationship between the alveolar macrophage and the septal fi broblast. Int J Radiat Oncol Biol Phys 24:93–101

Rubin P, Johnston CJ, Williams JP, et al. (1995) A perpetual cascade of cytokines post-irradiation leads to pulmonary fi brosis. Int J Radiat Oncol Biol Phys 33:99–109

Sause W, Kolesar P, Taylor S IV, et al. (2000) Final results of phase III trial in regionally advanced unresectable non- small cell lung cancer. Radiation Therapy Oncology Group, Eastern Cooperative Oncology Group, and Southwest Oncology Group. Chest 117:358–364

Schaake-Koning C, van den Bogaert W, Dalesio O, et al. (1992) Effects of concomitant cisplatin and radiotherapy on inop- erable non-small cell lung cancer. N Engl J Med 326:524–

530

Schiff PB, Horwitz SB (1980) Taxol stabilizes microtubules in mouse fi broblast cells. Proc Natl Acad Sci USA 77:1561–

1565

Segawa Y, Takigawa N, Kataoka M, et al. (1997) Risk factors for development of radiation pneumonitis following radiation therapy with or without chemotherapy for lung cancer. Int J Radiat Oncol Biol Phys 39:91–98

Sigel AV, Centrella M, Eghbali-Webb M (1996) Regulation of proliferative response of cardiac fi broblasts by trans- forming growth factor-beta 1. J Mol Cell Cardiol 28:1921–

1929

Sinclair WK, Morton RA (1966) X-ray sensitivity during the cell generation cycle of cultured Chinese hamster cells.

Radiat Res 29:450–474

Sleijfer S (2001) Bleomycin-induced pneumonitis. Chest 120:617–624

Steren A, Sevin B-U, Perras J, et al. (1993) Taxol sensitizes human ovarian cancer cells to radiation. Gynecol Oncol 48:252–258

Thomas AL, Cox G, Sharma RA, et al. (2000) Gemcitabine and paclitaxel associated pneumonitis in non-small cell lung cancer: report of a phase I/II dose escalating study. Eur J Cancer 36: 2329–2334

Tishler RB, Geard CR, Hall EJ, et al. (1992) Taxol sensitizes human astrocytoma cells to radiation. Cancer Res 52:3495–3497 Wang GS, Yang KY, Perng RP (2001) Life-threatening hyper-

sensitivity pneumonitis induced by docetaxel (Taxotere).

Br J Cancer 85:1247–1250

Yorke ED, Jackson A, Rosenzweig KE, et al. (2002) Dose- volume factors contributing to the incidence of radiation pneumonitis in non-small-cell lung cancer patients treated with three-dimensional conformal radiation therapy. Int J Radiat Oncol Biol Phys 54:329–339

Zhang D, Jacobberger JW (1996) TGF-ß1 perturbation of the fi broblasts cell cycle during exponential growth: switching between negative and positive regulation. Cell Proliferation 29:289–307

Riferimenti

Documenti correlati

Protein solubility is a key aspect for many biotechnological, biomedical and industrial pro- cesses, such as the production of active proteins and antibodies. In addition,

It then discusses the impact of cross-disciplinary research on comparative-law taxonomies (such as territorial constitution and litigation, territorial demarcation, and

Estimation of Fundamental Stellar Properties Fundamental stellar properties can be estimated by compar- ing global asteroseismic parameters and complementary spectroscopic

In the current version, TRONCO implements the CAPRESE and CAPRI algorithms for cancer progression inference together with functionalities to load input cross-sectional data, set up

Chemical modifications of Tonda Gentile Trilobata hazelnut and derived processing products under different infrared and hot-air roasting conditions: a combined analytical

3D treatment planning, conformal radiotherapy, with consideration of both external radiotherapy and brachytherapy, stereotactic radiotherapy, intensity-modulated radiation

A meta-analysis of several randomized clinical trials in the adjuvant set- ting demonstrated a 5% improvement in 5-year sur- vival rate and a 13% decrease in relative risk of death

Methodology: The studies included in this thesis comprise a narrative review of the existing literature about the involvement of β-AR on proliferation of human cancer