Clinical Experiences with Carbon Ion Therapy



Courtesy Prof. Dr. Gerhard Kraft





5.4 Carbon Ion Exploration in Future Clinical Trials


Currently, clinical trial design is based on the assumption that the same biological effective dose is administered by photons, protons, or carbon ions. However, it is necessary to equate these doses, breaking the use of the traditional reference to photon dose. Therefore, it is important to look at differences in RBE and tissue type in order to create the best therapeutic ratio.

The design of future clinical screenings should address the differences in radiosensitivity between radio-resistant cells for low-LET RT and sensitivity for carbon ion RT to enable proper selection of histologies and patients who may benefit from this modality based on biomarkers and imaging. Patient selection should follow the protocol outlined in figure [4], acknowledging that none of the individual measures, whether hypoxia, alpha/beta ratio, or tumor proliferation, may represent the tumor microenvironment and true radiosensitivity of the tumor.

A332412_1_En_5_Fig1_HTML.gif


Fig. 5.1
Grading scale of histologies to warrant carbon ion exploration in future clinical trials. Grading scale that should be used to select patients and tumor histologies to determine inclusion earlier or later in clinical trials. Courtesy Springer

In Fig. 5.1, a cumulative score greater than or equal to 5 describes a radio-resistant tumor for low LET irradiation with standard of care (SOC), causing significant toxicity. Tumor histology and patients with 0–1 score have significant benefits from SOC treatment. The use of carbon ion therapy is sensitive when the advantages of using carbon ions exceed the therapeutic advantages that can be obtained with fractionated photon RT. With the advent of personalized medicine, those tumors that respond well to other radiation species should continue to be treated with these species, while a rare malignancy or patients who do not respond should be treated with carbon ions in an individualized way.


5.5 Clinical Results


Carbon ion therapy has demonstrated benefits for the following:

1.

Adenocarcinoma

 

2.

Adenoid cystic carcinoma (ACC)

 

3.

Malignant melanoma

 

4.

Sarcomas arising in the head-neck and many other sites

 

5.

Chordomas of the skull base and sacrum: Significant improvements have been achieved with proton and carbon ion RT; in long-term observation (10 years), the difference in local control rates became larger for carbon ion therapy.

 

6.

Bone and soft tissue sarcomas, including osteosarcoma, chordoma and many other types of sarcomas rising from head and neck, pelvis, vertebra/paravertebral and retroperitoneal region: These tumors are difficult to treat with surgery and are generally photon-resistant.

 

7.

Postsurgical pelvic recurrence of rectal cancer: Treatment has shown comparable or even better results than those achieved with surgery.

 

8.

Malignant melanoma and cancer of pancreas: A combination of carbon ion radiotherapy and chemotherapy has prevented or delays the development of distant metastases with improved survival and local control.

 

With the unique properties of carbon ions, treatment can be completed in a shorter period of time and with smaller fractions. Future directions of carbon ion therapy depend on the interaction between radiobiology, radiation oncology, and physics accelerators, which combined with the clinical results make it a very promising technique.


5.6 Clinical Advantages of Carbon Ions


The clinical advantages of carbon ions include improved therapeutic gain, hypofractionated radiotherapy, and potential suppression of metastases, as discussed in the following sections.


5.6.1 Improved Therapeutic Gain


The RBE of high-LET carbon ions is greater than low-LET photons and protons. Radiobiological advantages are expected when using carbon ions, such as decreased radiation damage, suppressed tissue population, reduced OER, and reduced dependence on radiosensitivity cell cycle. These advantages are maximized at the peak region, which, combined with improved physical dose location, may have an important role in improving the therapeutic ratio of carbon ion beams and beams of protons and photons. The RBE of carbon ions is similar to that of fast neutron beams. If we apply to the beams of carbon ions findings to the fast neutron therapy [5], carbon ion therapy appears to be effective against photon-resistant tumors and those located near critical structures [6].


5.6.2 Hypofractionated Radiotherapy (Without Enhancing Toxicity)


Because of the physical and biological characteristics of carbon ion therapy, it is possible to perform a hypofractionated radiation in relation to the standard used for photons. Experiments with fast neutron beams have shown that increasing the dose per fraction tends to lower RBE for both tumors and normal tissues [7]; however, RBE for tumors does not decrease as rapidly as RBE for normal tissues. Thus, the therapeutic ratio would increase rather than decrease, even if the dose fraction was increased. Experiments conducted with carbon ions, [8, 9] show similar results; therefore, a hypofractionated scheme may be used in carbon ion therapy without increased toxicity.


5.6.3 Potential Suppression of Metastases


As is known, the irradiation of carbon ions induces DNA damage, resulting in a high breakage of double-strand DNA, possibly suppressing the metastasis ability of cancer cells in relation to irradiation by X-rays. This is an advantage of treatment using carbon ions. Further studies are needed to confirm these discoveries. Secondary cancer induction after carbon ion therapy, also remains to be studied because there are not valid clinical data.


5.7 The Risk of Secondary Malignancies


More than 38 centers worldwide have treated more than 100,000 cancer patients with particle therapy. Most patients have been treated using protontherapy, but the use of carbon ions is increasing. Unfortunately, despite recent advances in radiotherapy, there is still the risk of cancer arising in a location that was previously free of disease, caused by the treatment itself and not metastasis. There is always the possibility of developing cancer under these conditions because of secondary neutrons, which are inevitably produced in treatments involving particle beams [10]. Quantifying these risks requires a detailed knowledge of a range of parameters and a multidisciplinary team.

Traditional radiotherapy has been improved as a consequence of the development of IMRT, which has enabled improved targeting of conventional X-rays and a reduction in the radiation dose exposure of healthy normal tissue. It is the state of the art in photonics therapy. However, this technique is less effective than carbon ion therapy (as used at HIT). IMRT requires two to three times more monitoring units to deliver a specific radiation dose to the tumor target, when compared with conformal radiotherapy delivered in three dimensions (3D-CRT). Using IMRT instead of 3D-CRT increases the risk of developing secondary cancer by a factor of approximately 2. It is important to remember that particle therapy beams deposit most of their energy near the end of their tracks in the region of the Bragg peak. This peak is spread out to cover the entire tumor volume, and the dose beyond the tumor is lower than in photon therapy.

The neutrons produced during radiation therapy collide with protons in water and generate additional charged particles that can ionize the surrounding molecules. However, this problem can be addressed by using magnetically scanned beams rather than passively scattered beams. It is worth noting that the characteristics of cancer vary from organ to organ, and there is no evidence that the tumor dose–response curves are the same for different organs.

Ionizing radiation is recognized by the World Health Organization (WHO) as a carcinogen. In regions exposed to high doses, ionizing radiation directly kills cells in the field; however, the resulting tissue inflammation and DNA damage to cells in the normal tissue surrounding the tumor can promote cancer. In children, cancer is fortunately relatively rare; when it occurs, radiotherapy is used to treat children with lymphoma, leukemia, brain tumors, sarcomas, Wilm’s tumor, neuroblastoma, and liver cancer [11]. In the radiotherapy of pediatric patients, the primary concern is low-dose exposure to distal organs; for adults, high radiation doses induce inflammation.

To estimate the risk of developing cancer from protons, we must rely entirely on animal and in vitro cell experiments. Estimates of the RBE of neutrons are largely based on animal studies, although atomic bomb survivors have also been exposed to neutrons and some data are available. However, considerable uncertainty remains in predicting the late effects of heavy ions in humans. These ions are effective in inducing inflammation. In general, only the organs in the beam path are exposed to heavy ions, while the distal organs receive scattered neutrons and protons.

There is good epidemiological evidence that radiation therapy can contribute to the long-term survival of children with cancer, but it also causes a high incidence of secondary malignancy among survivors. However, the data suggest that hadron therapy leads to a reduced risk of secondary malignancy as compared with conventional radiotherapy modalities that employ X-rays. When using heavy ions, the radiation dose to healthy normal tissues is very low. In addition, the production of neutrons by these ions is lower than is the case for protons, because fewer ions than protons are needed to achieve the same dose in the tumor target.

The lack of resources on hadron therapy has led to inadequate recommendations of continued conventional radiotherapy in cases of cancer recurrence. This has prevented proper treatment with hadron therapy techniques using carbon ions, leading patients to undergo chemotherapy as the only treatment, which is not always satisfactory.


5.8 Clinical Trials at HIT


Fifteen trials were initiated at HIT since November 14, 2009, when the first patient was treated. A brief description of each trial follows [12]:

1.

HIT-1 trial for chordomas of the skull base The first trial at HIT was a prospective randomized phase II trial for the treatment of skull base chordomas. This trial tested for superiority of carbon ion irradiation against proton irradiation with respect to the local progression-free survival. The 5-year local progression-free survival was 70 % using protons and 80 % using carbon ions.

 

2.

HIT chondrosarcoma trial This trial is also a prospective randomized phase II trial for the optimal treatment of skull base chondrosarcomas.

 

3.

COSMIC trial The goal of the trial was to evaluate toxicity in dose-escalated treatment with intensity-modulated radiotherapy (IMRT) and carbon ion boost for malignant salivary gland tumors of the head and neck.

 

4.

ACCEPT trial The ACCEPT trial followed the COSMIC trial in the treatment of adenoid cystic carcinomas (ACC) of the head and neck. However, in this trial, only ACC patients with microscopic residual disease can be included.

 

5.

IMRT-HIT-SNT trial This trial examines the effect of a carbon ion boost in the treatment of patients with unresected or incompletely resected nasal or paranasal sinus carcinomas.

 

6.

TPF C-HIT trial Locally advanced tumors of the oropharynx, hypopharynx, and larynx are suitable for this trial. Except for the induction chemotherapy using docetaxel, cisplatinum and 5FU (TPF), the design is similar to the ACCEPT trial.

 

7.

CLEOPATRA trial This trial is a single-center randomized phase II trial for the treatment of glioblastomas. The aim of the trial is to show the overall survival (primary end point) for glioblastoma patients using a carbon ion boost dose escalation compared to the standard treatment.

 

8.

CINDERELLA trial The effect of carbon ion irradiation in the treatment of recurrent gliomas after initial radiation treatment is examined in the CINDERELLA trial.

 

9.

MARCIE trial Atypical meningiomas have a much higher recurrence rate than meningiomas of WHO grade I. The Phase II MARCIE study evaluates a carbon ion boost applied to the macroscopic tumor in conjunction with photon radiotherapy in patients with atypical meningiomas after incomplete resection or biopsy.

 

10.

IPI trial The role of the use of ions in the primary treatment of prostate cancer is unknown. There are no prospective proton data. However, NIRS has published promising results in respect of the hypofractionated use of carbon ions. The IPI trial wants to confirm these Japanese data in a prospective randomized phase II trial.

 

11.

PROLOG trial This trial focuses on the use of protons in the postoperative situation of prostate cancer (either as adjuvant treatment or as salvage treatment).

 

12.

PANDORA trial Patients with recurrent rectal cancer are still challenging. This trial examines the role of carbon ions in the reirradiation of patients with recurrent rectal cancer.

 

13.

ISAC trial Imai et al. published the NIRS data with respect to hypofrationated carbon ion irradiation of sacral chordoma. This trial will confirm these data using the raster scan method. Additionally, it will be examined if these results are an effect of the use of carbon ions or an effect of high and hypofractionation.

 

14.

OSCAR trial This trial is a non-randomized therapy trial to determine the safety and efficacy of heavy ion therapy in patients with nonresectable osteosarcomas. The primary endpoint of OSCAR is feasibility and toxicity in the ion treatment of unresectable osteosarcoma.

 

15.

PROMETHEUS trial The PROMETHEUS trial is the first trial evaluating carbon ion radiotherapy delivered by intensity-modulated raster scanning for the treatment of hepatocellular carcinoma.

 

Assays were initiated to assess the role of carbon ions and protons in the treatment of a variety of cancer types. In the future, these will be included in HIT trials with moving targets. For example, the INKA test will start neoadjuvant radiotherapy using raster scanned carbon ions in patients with locally advanced sulcus superior tumors. The role of ions for radiotherapy in the treatment of pancreatic cancer will be developed, as well as the use of carbon ions for inoperable esophageal cancer [12].


5.9 Consolidated, Prospective, and Exceptional Indications Using Carbon Ion Therapy



5.9.1 Consolidated Indications


Consolidated indications (Table 5.2) are the core indications that have been treated effectively using neutron therapy (salivary gland tumors, adenoid cystic carcinomas of the upper respiratory and digestive tracts, particularly the trachea, superficial sarcomas) and are currently treated in Japan and Germany (adenocarcinomas of the head and neck, mucosal melanomas, chordomas, sarcomas, hepatocellular carcinomas, pelvic recurrences of rectal adenocarcinomas). Their published outcomes are well above the figures obtained using non-carbon ion therapy (approximately 20–25 % higher for 5-year local control) [13].


Table 5.2
Consolidated indications resulting from Etoile’s work

















Tumor location

Detailed definition of indications

Recommended form of hadron therapy

Estimated incidencea (cases/year in France)

Salivary gland (parotid gland) tumours

Inoperable tumours or refusal of surgery or R2 restrictions or local recurrencesb

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 28, 2017 | Posted by in BIOCHEMISTRY | Comments Off on Clinical Experiences with Carbon Ion Therapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access