Skip to main content
Cancer Science logoLink to Cancer Science
. 2024 Jan 26;115(3):954–962. doi: 10.1111/cas.16065

Patient survey on cancer genomic medicine in Japan under the national health insurance system

Hidenori Kage 1,2, Nana Akiyama 1, Hyangri Chang 1, Aya Shinozaki‐Ushiku 1,3,4, Mirei Ka 3, Junichi Kawata 5, Manabu Muto 6, Yusuke Okuma 7, Natsuko Okita 7, Katsuya Tsuchihara 8, Junko Kikuchi 9, Hidekazu Shirota 10, Hideyuki Hayashi 11, Toshio Kokuryo 12, Shinichi Yachida 13, Akira Hirasawa 14, Makoto Kubo 15, Hirotsugu Kenmotsu 16, Masahiko Tanabe 17, Tetsuo Ushiku 4, Kaori Muto 5, Yasuyuki Seto 18, Katsutoshi Oda 1,3,
PMCID: PMC10920978  PMID: 38273803

Abstract

In Japan, comprehensive genomic profiling (CGP) tests have been reimbursed under the national health care system for solid cancer patients who have finished standard treatment. More than 50,000 patients have taken the test since June 2019. We performed a nation‐wide questionnaire survey between March 2021 and July 2022. Questionnaires were sent to 80 designated Cancer Genomic Medicine Hospitals. Of the 933 responses received, 370 (39.7%) were web based and 563 (60.3%) were paper based. Most patients (784, 84%) first learned about CGP tests from healthcare professionals, and 775 (83.1%) gave informed consent to their treating physician. At the time of informed consent, they were most worried about test results not leading to novel treatment (536, 57.4%). On a scale of 0–10, 702 respondents (75.2%) felt that the explanations of the test result were easy to understand (7 or higher). Ninety‐one patients (9.8%) started their recommended treatment. Many patients could not receive recommended treatment because no approved drugs or clinical trials were available (102/177, 57.6%). Ninety‐eight patients (10.5%) did not wish their findings to be disclosed. Overall satisfaction with the CGP test process was high, with 602 respondents (64.5%) giving a score of 7–10. The major reason for choosing 0–6 was that the CGP test result did not lead to new treatment (217/277, 78.3%). In conclusion, satisfaction with the CGP test process was high. Patients and family members need better access to information. More patients need to be treated with genomically matched therapy.

Keywords: cancer genomic medicine, comprehensive genomic profiling, national health care system, patient satisfaction, survey


In Japan, where comprehensive genomic profiling tests are given as part of the national healthcare system, patient satisfaction of the test process was high. More patients need to be treated with genomically matched therapy.

graphic file with name CAS-115-954-g002.jpg


Abbreviation

CGP,

comprehensive genomic profiling.

1. INTRODUCTION

Comprehensive cancer genomic profiling (CGP) tests are used worldwide with aims to improve the prognosis of advanced cancer patients. In Japan, CGP tests have been approved for reimbursement under the national health insurance, although the following conditions must be met: (1) the patient has a solid tumor in which standard therapy is expected to be completed or is unavailable; (2) the patient is well enough to receive treatment if it is recommended by a molecular tumor board, specifically called expert panels; (3) the CGP tests are done at designated core hospitals for cancer genomic medicine responsible for training and research in cancer genomic medicine and holding expert panels, designated hospitals responsible for holding expert panels, or cooperative hospitals, all of which are assigned by the Ministry of Health, Labour and Welfare 1 ; (4) a prespecified informed consent procedure is followed; (5) every test result must be discussed at expert panels; and (6) clinical and genomic data are submitted to the Center for Cancer Genomics and Advanced Therapeutics if patient consent is obtained. 2 , 3 As of March 2023, more than 50,000 advanced solid cancer patients have undergone CGP testing using the national health insurance system.

Taking a CGP test can benefit patients with advanced solid cancer through detection of somatic gene mutations leading to targeted therapy, diagnosis of cancer type, or detection of germline variants suggestive of hereditary cancer. However, while novel therapy is suggested to 44% of the patients, less than 10% subsequently receive it in Japan. 4 , 5 This means that while all patients take CGP tests at the end of standard therapy, if any, most find out that taking the test did not result in new treatment options. The limited access to relevant therapies negatively influences the psychology of both cancer patients and their families. 6 , 7 , 8 Disclosure of germline findings is also associated with anxieties of the patients and families. 9 , 10 Furthermore, knowledge about genomics and CGP tests is often limited for the patients. 9 , 11 , 12 However, surveys about the psychological impact of CGP on cancer patients and their relatives have been limited. 13 , 14 Considering the uniqueness of CGP tests in Japan, that is, (i) they are only applicable to cancer patients who have (almost) finished standardized therapies, and (ii) they are reimbursed under the national health insurance, it is very important to address the psychological effects of CGP tests at late lines of therapies.

Here, we conducted a survey to determine the level of patient satisfaction and concerns regarding the process of cancer genomic medicine.

2. MATERIALS AND METHODS

2.1. Institutions

Starting in March 2021, questionnaires were sent to all 12 core hospitals, 23 of 33 designated hospitals, and 45 of 189 cooperative hospitals. Initially, we asked each hospital to hand out an information sheet to each patient when the CGP test results were given to them, and the patients or family members responded to the questionnaire over the web using a QR code. Because patient response was limited, we added a paper‐based questionnaire starting in August 2021, and the patients could choose to respond over the web or on paper. Patients and/or family members responded without help from healthcare professionals. This is an interim analysis of the first 933 responses that we received through July 2022. This study was approved by the Research Ethics Committee of the Faculty of Medicine at The University of Tokyo (#2020378NI) and all institutional review boards at participating hospitals. Written or web‐based informed consent was obtained from all respondents.

2.2. Types of CGP tests

Three types of CGP tests were approved before (or during) the study period in Japan: a tumor/normal paired CGP test (OncoGuide™ NCC Oncopanel System), 15 a tumor‐only CGP test (FoundationOne®CDx), and a liquid biopsy CGP test (FoundationOne®Liquid CDx). 16

2.3. The questionnaire

We asked patients about their information sources, their level of comprehension, and their satisfaction. Most answers were to be given on a scale of 0–10, and 7 or higher was considered satisfactory while 6 or lower was considered unsatisfactory. The questionnaire was in Japanese, and the English translation is shown in Table S1.

2.4. Summary of free comments

The data were analyzed employing an inductive coding approach, using NVivo 12 (QSR International). N.A. performed the coding, and N.A. and H.C. verified the validity of each code.

2.5. Statistics

Categorical data were compared by the chi‐squared test. A p‐value of less than 0.05 was considered statistically significant. Data were analyzed with Sigma Plot version 12.3 (Systat Software).

3. RESULTS

3.1. Characteristics of the respondents

As of July 2022, we received 933 responses (Table 1), of which 370 (39.7%) were web based and 563 (60.3%) paper based. Respondents were patients (761, 81.6%), family members (152, 16.3%), or others (20, 2.1%). Patients in their 60s and 70s made up the largest group (26.8% and 29.6%, respectively), while those at the age of 90 and above and less than 10 were the smallest group (0.2% and 0.5%, respectively). Four hundred respondents (42.9%) identified the patients as female and 510 (54.7%) as male. Most patients (655, 70.2%) underwent CGP testing at their treating hospital, while 254 (27.2%) patients were referred to a Cancer Genomic Medicine Hospital to take the test. One hundred and thirty‐four patients (13.9%) took OncoGuide™ NCC Oncopanel, 525 (54.3%) took FoundationOne®CDx, 108 (11.2%) took a liquid‐based CGP test, and 151 (15.6%) did not know which test they took. From the time the patients requested a CGP test, 135 (14.5%) received the test result within 4 weeks, 645 (69.1%) within 8 weeks, and 814 (87.2%) within 12 weeks.

TABLE 1.

Characteristics of the respondents.

Number of respondents Percentage
Total number of respondents 933
Web based 370 39.7%
Paper based 546 58.5%
Other 17 1.8%
Respondents
Patients 761 81.6%
Family members 152 16.3%
Other 20 2.1%
Age
<10 5 0.5%
10–19 11 1.2%
20–29 7 0.8%
30–39 29 3.1%
40–49 107 11.5%
50–59 194 20.8%
60–69 250 26.8%
70–79 276 29.6%
80–89 33 3.5%
90 and above 2 0.2%
Sex
Male 510 54.7%
Female 400 42.9%
Other 23 2.5%
Place of testing
Treating hospital 655 70.2%
Referred for test 254 27.2%
Other 24 2.6%
Test
NCC Oncopanel 134 14.4%
FoundationOne® CDx 525 56.3%
Liquid biopsy 108 11.6%
Other 30 3.1%
Unknown 151 16.2%
No response 18 1.9%
Time between requesting a test and receiving a result
<4 weeks 135 14.5%
4–6 weeks 273 29.3%
6–8 weeks 237 25.4%
8–10 weeks 117 12.5%
10–12 weeks 52 5.6%
12 weeks and longer 82 8.8%
No response 37 4.0%

3.2. Source of information

Physicians and other healthcare professionals were the main sources of information (784, 84%), while family members, friends, and websites were each less than 3% (Table 2). Most respondents found physicians and other health care professionals to be highly reliable (607, 65.1%) or somewhat reliable (286, 30.7%) sources of information.

TABLE 2.

Main source of information for comprehensive genomic profiling.

Source of information Number of patients Percentage
Physicians and healthcare professionals 784 84.0
Family members 16 1.7
Friends or colleagues 9 1.0
Websites and social network services 52 5.6
Others 72 7.7

3.3. Satisfaction with consultation and informed consent

Many patients consulted a health care professional before consenting to take a CGP test (693 patients [74.3%] consulted vs. 222 patients [23.8%] did not consult): 665 (96.0%) patients consulted their treating physician, 92 (13.3%) consulted another physician, 70 (10.1%) consulted a cancer genomic medicine coordinator (a nonphysician healthcare professional who was trained by a certified CGP program), and 42 (6.1%) consulted a nurse (Figure 1A). 85.3%, 82.6%, 90.3%, and 88.6% of the respondents who consulted their treating physician, another physician, a cancer genomic medicine coordinator, or a nurse, respectively, responded as satisfied or somewhat satisfied are shown in Table 3.

FIGURE 1.

FIGURE 1

Satisfaction of consultation and informed consent. (A) The percentage of healthcare professionals patients consulted before informed consent. (B) The percentage of healthcare professionals patients encountered for informed consent. (C) Level of satisfaction toward physicians during informed consent. (D) Level of satisfaction towards nonphysician healthcare professional during informed consent. (E) Patient worries at the time of informed consent.

TABLE 3.

The reasons for choosing satisfied or somewhat satisfied while consulting a healthcare professional over comprehensive genomic profiling tests (multiple answers allowed).

Treating physician Other physician Cancer genomic medicine coordinator Nurse
Easy access 74 (13.1%) 10 (13.2%) 7 (13.8%) 9 (17.9%)
Spent enough time 105 (18.6%) 20 (26.3%) 21 (32.3%) 10 (25.6%)
Encounter was helpful 187 (33.1%) 31 (40.8%) 24 (36.9%) 23 (59.0%)
Explanation was easy to understand 286 (50.6%) 58 (76.3%) 40 (61.5%) 12 (30.8%)
Other 26 (4.6%) 3 (3.9%) 6 (9.2%) 1 (2.6%)

Note: p = 0.09.

All patients gave informed consent to undergo a CGP test. Most patients (775, 83.1%) discussed the matter with their treating physician, 195 (20.9%) with another physician, 106 (11.4%) with a cancer genomic medicine coordinator, and 97 (10.4%) with a nurse; 99 patients (10.6%) did not recall the occupation of the health care professional (Figure 1B). The levels of satisfaction (on a scale of 0–10) toward physicians and nonphysician healthcare professionals during the process of informed consent for the CGP test are shown in Figure 1C,D. When patients took the CGP test, they were most worried about test results not leading to a novel treatment (536, 57.4%), the cost of the CGP tests (269, 28.8%), the cost of the novel treatment (226, 24.2%), and the amount of time before test results are returned (205, 22.0%) (Figure 1E).

3.4. Recommendation of treatment

On a scale of 0–10, 702 respondents (75.2%) felt that the explanations of the test result were easy to understand (7 or higher), and 210 (22.5%) felt the explanations were hard to understand (6 or lower) (Figure 2A). The reasons for the 702 responses with high satisfaction were: explanation in plain language (440, 62.7%), taking plenty of time to explain the results (330, 47.0%), and being able to ask questions (365, 52.0%) (Figure 2B). Treatment was recommended based on test results in 437 patients (46.8%), and no treatment was recommended in 473 patients (50.7%) (Figure 2D). Ninety‐one patients (9.8% of all patients) started their recommended treatment, 177 (40.5%) did not receive recommended treatment, and 194 (44.4%) were undecided. Of the 91 patients, 64 (70.3%) received approved drugs, and 15 (16.5%) participated in a clinical trial. The reason 177 patients did not receive the recommended treatment was that no approved drugs or clinical trials were available in 102 patients (57.6%) and 30 patients (16.9%) did not meet clinical trial criteria (Figure 2E).

FIGURE 2.

FIGURE 2

Satisfaction during explanation of test results and recommendation of treatment. (A) Level of difficulty in understanding test results. (B) The reasons respondents chose 7–10 in graph (A). (C) The reasons respondents chose 0–6 in graph (A). (D) The percentage of patients who received treatment recommendations and started treatment. (E) The reasons patients could not start their treatment despite receiving a treatment recommendation.

3.5. Germline findings

For germline findings, 722 patients (77.4%) requested that their findings be disclosed, while 98 patients (10.5%) did not wish their findings to be disclosed (Figure 3A). Some patients did not want their germline findings to be disclosed because they did not think it would affect their treatment choices (37/98, 37.8%) and they were worried about the impact on their family members (20/98, 20.4%) (Figure 3B). Of the 722 patients who wished for germline disclosure, 166 (23.0%) found they had a (presumed) pathogenic germline variant, and 520 (72.0%) did not. Of the 166 patients who had a (presumed) germline variant disclosed, 132 (79.5%) responded that they received adequate or somewhat adequate genetic counseling (Figure 3C). Out of 154 responses, 131 (85.1%) gave a score of 7–10 and found that the explanations were easy to understand, while 23 (14.9%) gave a score of 3–6 (Figure 3D). The reasons 131 patients gave a high score of 7–10 were: explaining in plain language (80, 61.1%), taking time to explain the results (69, 52.7%), and being able to ask questions (66, 50.4%) (Figure 3E).

FIGURE 3.

FIGURE 3

Responses on germline findings. (A) The percentage of patients asking for disclosure of germline findings. (B) The reasons patients did not want their germline findings disclosed. (C) Satisfaction of genetic counseling. (D) Level of difficulty in understanding test results. (E) The reasons respondents chose 7–10 in graph (D).

3.6. Overall satisfaction

Overall satisfaction toward support by the hospital was high, with 606 respondents (65.0%) giving a score of 7–10 and 261 respondents (28.0%) giving a score of 0–6 (Figure 4A). Most respondents answered that support was helpful (460/606, 75.9%), but many also thought it was hard to find support (117/261, 44.8%) (Figure 4B,C). Overall satisfaction toward the CGP test process was high, with 602 respondents (64.5%) giving a score of 7–10 and 277 respondents (29.7%) giving a score of 0–6 (Figure 4D). The reasons for a score of 7–10 were that respondents could consider the cancer risk of family members (273/602, 45.3%), the test results were explained in detail (255/602, 42.4%), the explanations were adequate (238/602, 39.5%), and the respondents could learn about their cancer (206/602, 34.2%) (Figure 4E). The major reason for choosing 0–6 was that taking the CGP test did not lead to new treatment (217/277, 78.3%) (Figure 4F).

FIGURE 4.

FIGURE 4

Satisfaction of hospital support and overall satisfaction toward cancer genomic medicine. (A) Level of satisfaction with hospital support. (B) The reasons respondents chose 7–10 in graph (A). (C) The reasons respondents chose 0–6 in graph (A). (D) Level of overall satisfaction with cancer genomic medicine. (E) The reasons respondents chose 7–10 in graph (D). (F) The reasons respondents chose 0–6 in graph (D).

3.7. Free comments

At the end of the survey, we included a comment section where respondents could freely write whatever they had in mind. Three hundred respondents wrote comments, including 69 comments (23%) on treatment, 67 comments (22%) on explanation of tests, 40 comments (13%) on cost, 37 comments (12%) on CGP tests, and 36 comments (12%) on the availability of CGP tests (Data S1).

4. DISCUSSION

To our knowledge, this is the first nationwide survey that asked patients about their experience with cancer genomic medicine under universal health insurance coverage. Because (1) cancer genomic medicine is a new field, (2) patients can take a CGP test only when standard treatment is finished, and (3) many CGP tests do not result in new treatment options, we considered it important to assess the patients' perspective on these tests. We found that patients were generally satisfied with the process of CGP test taking, but concerns for each step also became apparent.

We found that respondents were younger than general cancer patients. According to cancer statistics, the percentages of cancer patients from under the age of 10 to above 80 by 10‐year intervals were 0.04%, 0.05%, 0.1%, 0.4%, 1.7%, 4.9%, 13.0%, 31.4%, and 48.4%, respectively. 4 This was expected because younger patients generally seek novel treatment and older patients often cannot participate in clinical trials. The male‐to‐female ratio was well balanced. In Japan, cancer deaths were seen in 222,467 males and 159,038 females in 2021, which was similar to the 510 to 400 ratio seen in our survey. 4 As expected, most patients underwent CGP testing at their treating hospital, as CGP tests are more accessible there. Conversely, this means we need to find better ways to deliver CGP tests to cancer patients not treated at a cancer genomic medicine hospital. Approximately 80% of the patients knew which CGP test they took, and 15% did not; this may be a surrogate for patient understanding of the test and seems satisfactory, but more can be done. The time between the patients requesting and receiving results for a CGP test was reasonable.

We found that the source of CGP test information was skewed toward physicians and other healthcare professionals and that most patients and family members did not receive information from other family members, friends, websites, or social network services. CGP test information is available on public websites such as that of the National Cancer Center “Cancer Information Japan” and Center for Cancer Genomics and Advanced Genomics, but they need more recognition.

As regards physicians and other healthcare professionals, consultation before informed consent was mostly done by physicians. If patients can consult other healthcare professionals, such as Cancer Genomic Medicine Coordinators and nurses, more patients may find themselves eligible for CGP tests. The process of informed consent was mostly satisfactory but with room for improvement. Patients mostly worried about test results not leading to new treatment, but cost was also an issue. The cost of a CGP test is fixed at ¥560,000 (¥560,000 = US$4000 at ¥140/US$) in Japan, and 70%–90% is reimbursed. 17 Further, patients are exempted from paying more than a fixed amount, which is calculated based on the patient's income. 18 In addition to increasing the number of patients who receive recommended treatment, both lowering payment and sufficient knowledge about the medical expense benefit system are important.

Respondents also found the explanation of test results to be mostly satisfactory. Less than 10% of the patients received recommended treatment, which is similar to what has been reported. 4 , 5 Of the patients who received treatment, most received approved drugs, and only a minority participated in a clinical trial. This is unexpected, as CGP tests are given only to patients who have finished standard treatment, which means that treatment based on genomic alterations should have been given already through the use of companion diagnostics. Our surprising finding suggests that (1) CGP tests are sometimes the only approved companion diagnostic (e.g., tumor mutational burden‐high/microsatellite instability‐high and pembrolizumab treatment, and FGFR and NTRK gene fusions and the respective inhibitors), (2) CGP tests may result in recommending treatments that are not standard but considered to have a high probability of being effective (e.g., use of platinum‐based chemotherapy in BRCA1/2‐altered pancreatic cancer), and (3) treating physicians may sometimes be undertreating patients. This also suggests that if patients underwent a CGP test before standard treatment is finished, these approved drugs may be used earlier, which can lead to better outcomes. Many patients did not receive the recommended treatment mostly because no approved drugs or clinical trials were available any longer. Indeed, first‐line CGP tests by FoundationOne®CDx in previously untreated advanced cancers increased the probability of treatment with a recommended drug to 19.8%. 19 Improvement in CGP panels, earlier implementation of CGP panels, and increasing the number of clinical trials are all likely necessary so that more patients can receive a novel treatment.

Ten percent of the patients did not wish their germline findings to be disclosed because they thought it does not affect their treatment choices and worried about the impact on family members. This suggests that patients may have declined disclosure without fully understanding the consequences. Germline findings are often associated with promising treatment choices, such as BRCA1/2 variants and PARP inhibitors and variants in mismatch repair genes and immune checkpoint inhibitors. Germline findings are also detected in a number of “off‐tumor” contexts, and the CGP tests are expected to complement conventional approaches to germline testing alone. 20 , 21 Because the difference between somatic and germline variants can be difficult to understand, a more thorough explanation may be needed in some patients. Those who received genetic counseling generally gave a high satisfaction score.

Many respondents thought it was hard to find hospital support while navigating the CGP test, and hospitals need to (1) improve the way they support cancer patients and (2) better promote the support system to patients. In addition, extensive efforts by physicians and other healthcare professionals are necessary at each hospital to complete all the processes of a CGP test in Japan. 17 Therefore, shifting the effort from the procedure of the tests to the support of patients is highly warranted. Finally, overall satisfaction toward the CGP test was high, and the reasons for a high score were multiple. Surprisingly, many patients who could not receive recommended therapies gave high scores for overall satisfaction. The reasons, except for receipt of new treatments, were mostly associated with gaining knowledge about their own cancers. As communications and knowledge about their own cancers are important to patients and family members throughout the process of cancer treatment, earlier CGP testing may help patients understand basic knowledge about their cancers, including hereditary cancer risks, which may increase their levels of satisfaction. Conversely, the major reason for choosing a low score was that taking a CGP test did not lead to new treatment. Again, delivery of novel treatment to these patients is urgently needed.

As this was a survey, some biases exist. First, the respondents to this questionnaire underwent CGP testing, and the opinion of cancer patients who did not take a CGP test is unknown. Additionally, the satisfaction scores of the respondents may have been generally high simply because they were able to take the test. The percentage of patients with germline findings was higher than generally reported. These patients usually have multiple visits from multiple healthcare workers, which may increase their level of satisfaction; however, the number was still small and likely did not skew the data significantly.

In conclusion, this is the first nationwide survey asking patients or their family members their opinions on CGP tests. Satisfaction was generally high, but many key issues were addressed. In particular, patients and family members need better access to information on CGP tests, and measures should be taken to enable more patients to receive a matched therapy after taking a CGP test.

AUTHOR CONTRIBUTIONS

Hidenori Kage: Conceptualization; data curation; formal analysis; investigation; methodology; visualization; writing – original draft; writing – review and editing. Nana Akiyama: Data curation; formal analysis; investigation; methodology; writing – review and editing. Hyangri Chang: Data curation; formal analysis; investigation; methodology; writing – review and editing. Aya Shinozaki‐Ushiku: Data curation; formal analysis; writing – review and editing. Mirei Ka: Data curation; writing – review and editing. Junichi Kawata: Data curation; formal analysis; investigation; writing – review and editing. Manabu Muto: Data curation; formal analysis; supervision; writing – review and editing. Yusuke Okuma: Data curation; formal analysis; writing – review and editing. Natsuko Okita: Data curation; formal analysis; writing – review and editing. Katsuya Tsuchihara: Data curation; formal analysis; writing – review and editing. Junko Kikuchi: Data curation; formal analysis; writing – review and editing. Hidekazu Shirota: Data curation; formal analysis; writing – review and editing. Hideyuki Hayashi: Data curation; formal analysis; writing – review and editing. Toshio Kokuryo: Data curation; formal analysis; writing – review and editing. Shinichi Yachida: Data curation; formal analysis; writing – review and editing. Akira Hirasawa: Data curation; formal analysis; writing – review and editing. Makoto Kubo: Data curation; formal analysis; writing – review and editing. Hirotsugu Kenmotsu: Data curation; formal analysis; writing – review and editing. Masahiko Tanabe: Data curation; formal analysis; writing – review and editing. Tetsuo Ushiku: Data curation; formal analysis; writing – review and editing. Kaori Muto: Data curation; formal analysis; project administration; writing – review and editing. Yasuyuki Seto: Funding acquisition; project administration; supervision; writing – review and editing. Katsutoshi Oda: Conceptualization; data curation; formal analysis; funding acquisition; investigation; methodology; project administration; supervision; validation; writing – review and editing.

CONFLICT OF INTEREST STATEMENT

H. Kage received a research fund from Konica Minolta, Inc. M. Muto received lecture fees from Chugai Pharmaceutical Co and MSD and a research fund from Chugai Pharmaceutical Co. Y. Okuma received research funds and lecture fees from Chugai Pharmaceutical Co. K. Tsuchihara received lecture fees from Chugai Pharmaceutical Co. A. Hirasawa received lecture fees from Chugai Pharmaceutical Co. H. Kenmotsu received lecture fees from Chugai Pharmaceutical Co. K. Oda received research funds from Konica Minolta, Inc. and Chugai Pharmaceutical Co. and lecture fees from Chugai Pharmaceutical Co. K. Tsuchihara, S. Yachida, and K. Oda are associate editors for Cancer Science. Other authors do not have any conflict of interest.

FUNDING INFORMATION

This study was supported by a grant from the Ministry of Health, Labour and Welfare (20EA1006).

ETHICS STATEMENTS

Approval of the research protocol by an Institutional Review Board: # 2020378NI.

Informed Consent: Written or web‐based informed consent was obtained from all respondents.

Registry and the Registration No. of the study/trial: jRCT1030210547.

Animal Studies: N/A.

Supporting information

Table S1.

CAS-115-954-s002.xlsx (25.7KB, xlsx)

Data S1.

CAS-115-954-s001.docx (20.5KB, docx)

ACKNOWLEDGMENTS

We would like to thank all the hospitals that participated in this study. We would like to thank the members of the Informed Consent Working Group, Secondary Findings Working Group, Repository Working Group, Expert Panel Working Group, Drug Discovery Working Group, and Dr. Takayuki Yoshino for their suggestions.

Kage H, Akiyama N, Chang H, et al. Patient survey on cancer genomic medicine in Japan under the national health insurance system. Cancer Sci. 2024;115:954‐962. doi: 10.1111/cas.16065

REFERENCES

  • 1. Mukai Y, Ueno H. Establishment and implementation of cancer genomic medicine in Japan. Cancer Sci. 2021;112:970‐977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Kohno T, Kato M, Kohsaka S, et al. C‐CAT: the National Datacenter for cancer genomic medicine in Japan. Cancer Discov. 2022;12:2509‐2515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Mano H. Cancer genomic medicine in Japan. Proc Jpn Acad Ser B Phys Biol Sci. 2020;96:316‐321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Cancer Information Service NCC, Japan . Cancer Statistics. (Vital Statistics of Japan, Ministry of Health, Labour and Welfare). 2021.
  • 5. Sunami K, Naito Y, Komine K, et al. Chronological improvement in precision oncology implementation in Japan. Cancer Sci. 2022;113:3995‐4000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Miller FA, Hayeems RZ, Bytautas JP, et al. Testing personalized medicine: patient and physician expectations of next‐generation genomic sequencing in late‐stage cancer care. Eur J Hum Genet. 2014;22:391‐395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Butow PN, Best MC, Davies G, et al. Psychological impact of comprehensive tumor genomic profiling results for advanced cancer patients. Patient Educ Couns. 2022;105:2206‐2216. [DOI] [PubMed] [Google Scholar]
  • 8. Nishino M, Fujimori M, Koyama T, et al. Prevalence of psychological distress, quality of life, and satisfaction among patients and family members following comprehensive genomic profiling testing: protocol of the quality of life for cancer genomics and advanced therapeutics (Q‐CAT) study. PloS One. 2023;18:e0283968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Nagai A, Ri I, Muto K. Attitudes toward genomic tumor profiling tests in Japan: patients, family members, and the public. J Hum Genet. 2019;64:481‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Hamilton JG, Shah IH, Salafia C, et al. Development of a novel measure of advanced cancer patients' perceived utility of secondary germline findings from tumor genomic profiling. PEC Innov. 2023;2:100124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Best MC, Bartley N, Jacobs C, et al. Patient perspectives on molecular tumor profiling: "why wouldn't you?". BMC Cancer. 2019;19:753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Wing SE, Hu H, Lopez L, et al. Recall of genomic testing results among patients with cancer. Oncologist. 2021;26:e2302‐e2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bartley N, Napier CE, Butt Z, et al. Cancer patient experience of uncertainty while waiting for genome sequencing results. Front Psychol. 2021;12:647502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Meiser B, Butow P, Davies G, et al. Psychological predictors of advanced cancer patients' preferences for return of results from comprehensive tumor genomic profiling. Am J Med Genet A. 2022;188:725‐734. [DOI] [PubMed] [Google Scholar]
  • 15. Sunami K, Ichikawa H, Kubo T, et al. Feasibility and utility of a panel testing for 114 cancer‐associated genes in a clinical setting: a hospital‐based study. Cancer Sci. 2019;110:1480‐1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Kanai M. Current clinical practice of precision medicine using comprehensive genomic profiling tests in biliary tract cancer in Japan. Curr Oncol. 2022;29:7272‐7284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kage H, Oda K, Muto M, et al. Human resources for administrative work to carry out a comprehensive genomic profiling test in Japan. Cancer Sci. 2023;114:3041‐3049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Ureshino H, Kamachi K, Kimura S. Japan's high‐cost medical expense benefit system could be an obstacle to promoting generic imatinib. Lancet Haematol. 2019;6:e498. [DOI] [PubMed] [Google Scholar]
  • 19. Matsubara J, Mukai K, Kondo T, et al. First‐line genomic profiling in previously untreated advanced solid tumors for identification of targeted therapy opportunities. JAMA Netw Open. 2023;6:e2323336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Tung N, Dougherty KC, Gatof ES, et al. Potential pathogenic germline variant reporting from tumor comprehensive genomic profiling complements classic approaches to germline testing. NPJ Precis Oncol. 2023;7:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Kuzbari Z, Bandlamudi C, Loveday C, et al. Germline‐focused analysis of tumour‐detected variants in 49,264 cancer patients: ESMO precision medicine working group recommendations. Ann Oncol. 2023;34:215‐227. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1.

CAS-115-954-s002.xlsx (25.7KB, xlsx)

Data S1.

CAS-115-954-s001.docx (20.5KB, docx)

Articles from Cancer Science are provided here courtesy of Wiley

RESOURCES