cancer grand challenges: embarking on a new era of discovery · 2020. 12. 8. · views january 2021...

6
VIEWS JANUARY 2021 CANCER DISCOVERY | OF1 IN FOCUS Cancer Grand Challenges: Embarking on a New Era of Discovery Iain Foulkes 1 and Norman E. Sharpless 2 1 Cancer Research UK, London, United Kingdom. 2 National Cancer Institute, Bethesda, Maryland. Corresponding Author: Norman E. Sharpless, National Cancer Institute, Bethesda, MD 20814. Phone: 240-781-3300; E-mail: norman.sharpless@ nih.gov Cancer Discov 2021;11:15 doi: 10.1158/2159-8290.CD-20-1657 ©2020 American Association for Cancer Research. Summary: Cancer Grand Challenges is a unique funding platform that dares global, multidisciplinary teams of researchers to come together, think differently, and tackle some of the toughest challenges in cancer research. Here, we discuss the nine intractable challenges currently open for application. INTRODUCTION With the development and approval of many new therapies based on a deep biological understanding of cancer, recent years have seen significant progress against the disease, including for cancers that were previously hard to treat. These hard-won gains, enabled by important insights from decades of science, have taught us a great deal—about cancer and about cancer research. Despite these advances, some difficult problems remain. These range from gaps in our fundamental understanding of basic biological processes and phenomena which have con- founded us for years, such as tumor cell dormancy, to global public health issues where evidence is limited and fragmented. Cancer Grand Challenges is a new global research plat- form to address these difficult but critical challenges in cancer research. Uniting Cancer Research UK and the U.S. National Cancer Institute, the initiative supports multi- disciplinary science on a global scale. By leveraging our combined resources and building on a global foundation of excellence in cancer research, together we can go further than we could independently. Together, we recently launched nine new challenges, invit- ing global, multidisciplinary teams to apply. Some, such as the “Inflammation” challenge, look to answer questions that have impeded progress for many years. Others, such as the “Normal Phenotypes” challenge, seek to capitalize on rela- tively recent but significant observations that could unlock new thinking about how cancer develops. Below, we set out the nine new challenges. By driving progress on all nine through the power of global scientific collaboration, Cancer Grand Challenges offers the potential to benefit entire scientific fields. EXTRACHROMOSOMAL DNA The idea that genes can be found on extrachromosomal DNA (ecDNA) is not new, but our appreciation of its impor- tance in cancer is growing. Although ecDNA was discovered in 1965, it took more than a decade for Alt and colleagues to move the field beyond phenomenology, with their discovery that tumor cells could develop resistance to methotrexate via the amplification of the DHFR gene on ecDNA elements (1). Oncogenes were reported to reside on ecDNA in the 1980s (2), and oncogene amplification was then shown to occur on ecDNA, the frequency and amplitude of which can be altered by different stimuli. Jump forward to the application of high-throughput sequencing, and we now know that onco- gene amplification on ecDNA occurs frequently, increasing extreme copy-number variation due to the unequal segrega- tion of circular amplicons, thereby accelerating intratumoral heterogeneity (3). Although it is becoming increasingly clear that ecDNA promotes aggressive tumor behavior, therapy resistance, and poorer survival, gaps in our knowledge about the origins, evo- lution, genomic organization, and clinical impact of ecDNA in human cancer remain. Therefore, this Cancer Grand Chal- lenge represents an opportunity to enhance our fundamental understanding of ecDNA—its formation, its role in tumor evolution and clonal dynamics, how it mediates drug resist- ance, and whether it can be targeted, which will be crucial to address its clinical implications. SENESCENCE Hayflick first described the limited replicative capacity of cells grown in vitro more than 50 years ago (4). Since this seminal discovery, the field of senescence has grown, with many ongoing efforts investigating its role in development, homeostasis, aging, and cancer. However, studying senescence in living organisms has been challenging due to the difficulty in identifying and charac- terizing senescent cells in tissues and organs; as yet, a gold- standard biomarker of the senescent state (in cancer cells) remains to be defined. We have, however, started to learn much from modeling studies. For example, experiments using the INK-ATTAC transgenic mouse model, generated by the van Deursen group, demonstrated how organism-wide elimination of senescent cells can extend the healthy life span and reduce the impact of age-related disorders, such as lordokyphosis and cataracts (5). Demaria and colleagues generated the p16- 3MR mouse, with which they confirmed the positive role of senescent cells in wound repair but the negative effects Research. on July 13, 2021. © 2020 American Association for Cancer cancerdiscovery.aacrjournals.org Downloaded from Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657

Upload: others

Post on 21-Feb-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cancer Grand Challenges: Embarking on a New Era of Discovery · 2020. 12. 8. · VIEWS January 2021 CANCER DISCOVERY | OF1 In Focus Cancer Grand Challenges: Embarking on a New Era

views

January 2021 CANCER DISCOVERY | OF1

In Focus

Cancer Grand Challenges: Embarking on a New Era of Discovery Iain Foulkes1 and Norman E. Sharpless2

1Cancer Research UK, London, United Kingdom. 2National Cancer Institute, Bethesda, Maryland.Corresponding Author: Norman E. Sharpless, National Cancer Institute, Bethesda, MD 20814. Phone: 240-781-3300; E-mail: [email protected] Discov 2021;11:1–5doi: 10.1158/2159-8290.CD-20-1657©2020 American Association for Cancer Research.

Summary: Cancer Grand Challenges is a unique funding platform that dares global, multidisciplinary teams of researchers to come together, think differently, and tackle some of the toughest challenges in cancer research. Here, we discuss the nine intractable challenges currently open for application.

IntroductIonWith the development and approval of many new therapies

based on a deep biological understanding of cancer, recent years have seen significant progress against the disease, including for cancers that were previously hard to treat. These hard-won gains, enabled by important insights from decades of science, have taught us a great deal—about cancer and about cancer research.

Despite these advances, some difficult problems remain. These range from gaps in our fundamental understanding of basic biological processes and phenomena which have con-founded us for years, such as tumor cell dormancy, to global public health issues where evidence is limited and fragmented.

Cancer Grand Challenges is a new global research plat-form to address these difficult but critical challenges in cancer research. Uniting Cancer Research UK and the U.S. National Cancer Institute, the initiative supports multi-disciplinary science on a global scale. By leveraging our combined resources and building on a global foundation of excellence in cancer research, together we can go further than we could independently.

Together, we recently launched nine new challenges, invit-ing global, multidisciplinary teams to apply. Some, such as the “Inflammation” challenge, look to answer questions that have impeded progress for many years. Others, such as the “Normal Phenotypes” challenge, seek to capitalize on rela-tively recent but significant observations that could unlock new thinking about how cancer develops.

Below, we set out the nine new challenges. By driving progress on all nine through the power of global scientific collaboration, Cancer Grand Challenges offers the potential to benefit entire scientific fields.

Extrachromosomal dnaThe idea that genes can be found on extrachromosomal

DNA (ecDNA) is not new, but our appreciation of its impor-tance in cancer is growing.

Although ecDNA was discovered in 1965, it took more than a decade for Alt and colleagues to move the field beyond phenomenology, with their discovery that tumor cells could develop resistance to methotrexate via the amplification of the DHFR gene on ecDNA elements (1).

Oncogenes were reported to reside on ecDNA in the 1980s (2), and oncogene amplification was then shown to occur on ecDNA, the frequency and amplitude of which can be altered by different stimuli. Jump forward to the application of high-throughput sequencing, and we now know that onco-gene amplification on ecDNA occurs frequently, increasing extreme copy-number variation due to the unequal segrega-tion of circular amplicons, thereby accelerating intratumoral heterogeneity (3).

Although it is becoming increasingly clear that ecDNA promotes aggressive tumor behavior, therapy resistance, and poorer survival, gaps in our knowledge about the origins, evo-lution, genomic organization, and clinical impact of ecDNA in human cancer remain. Therefore, this Cancer Grand Chal-lenge represents an opportunity to enhance our fundamental understanding of ecDNA—its formation, its role in tumor evolution and clonal dynamics, how it mediates drug resist-ance, and whether it can be targeted, which will be crucial to address its clinical implications.

sEnEscEncEHayflick first described the limited replicative capacity

of cells grown in vitro more than 50 years ago (4). Since this seminal discovery, the field of senescence has grown, with many ongoing efforts investigating its role in development, homeostasis, aging, and cancer.

However, studying senescence in living organisms has been challenging due to the difficulty in identifying and charac-terizing senescent cells in tissues and organs; as yet, a gold-standard biomarker of the senescent state (in cancer cells) remains to be defined.

We have, however, started to learn much from modeling studies. For example, experiments using the INK-ATTAC transgenic mouse model, generated by the van Deursen group, demonstrated how organism-wide elimination of senescent cells can extend the healthy life span and reduce the impact of age-related disorders, such as lordokyphosis and cataracts (5). Demaria and colleagues generated the p16-3MR mouse, with which they confirmed the positive role of senescent cells in wound repair but the negative effects

Research. on July 13, 2021. © 2020 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657

Page 2: Cancer Grand Challenges: Embarking on a New Era of Discovery · 2020. 12. 8. · VIEWS January 2021 CANCER DISCOVERY | OF1 In Focus Cancer Grand Challenges: Embarking on a New Era

Views

OF2 | CANCER DISCOVERY January 2021 AACRJournals.org

of chemotherapy-induced senescence in normal tissues (6). More recently, the Bernards group demonstrated the power of a one–two punch method to induce and selectively elimi-nate senescent cancer cells (senolysis) in hard-to-treat hepa-tocellular cancer (7).

These studies provide important insight about the thera-peutic potential of perturbing senescence, but we need to understand more to realize this potential. A key element of this Cancer Grand Challenge is establishing reliable bio-markers to identify senescent cancer cells to begin to answer important questions about senescence. The challenge also seeks to understand the physiologic impact of inducing senescence and/or removing senescent cells in the wider con-text of tumor development, and to identify ways to selectively kill senescent cancer cells.

E-cIgarEttEsDespite the increased use of electronic cigarettes (e-ciga-

rettes) over the past decade, including among children and young adults, we do not fully understand the health conse-quences of their use and whether they are effective at helping adult smokers to quit.

The strength of the evidence regarding e-cigarettes as a tool to aid smoking cessation is currently assessed as very low to moderate, according to systematic reviews employing GRADE (Grading of Recommendations, Assessment, Devel-opment and Evaluations) criteria for assessing certainty of the evidence. This is attributed to the limited number of randomized trials, heterogeneity across studies, methodologic and sampling limitations, and imprecision of results—both statistically and due to a small number of relevant studies. Moreover, few studies account for important variables related to characteristics of e-cigarette products used (e.g., nicotine content, voltage settings) and patterns of use (e.g., frequency, intensity; refs. 8, 9).

Furthermore, the evidence is still emerging regarding the short- and long-term health harms of e-cigarette use and how these products affect tobacco use patterns, including initia-tion, dual use of e-cigarettes and cigarettes, and smoking ces-sation (10). Toxicant levels are generally lower in e-cigarette aerosol than in cigarette smoke. Although the current body of research remains limited, research in cell systems, ani-mals, and humans demonstrates that e-cigarettes and their associated chemicals can have adverse effects on multiple organ systems, including the cardiovascular, respiratory, and immune systems (11). Moreover, youth e-cigarette use presents increased risk of health harms, including nicotine addiction, harm to the developing brain, and increased risk of cigarette smoking (10, 12).

Carefully designed and rigorous studies are needed to determine the harms of e-cigarette use and any potential benefits. Multinational and multidisciplinary approaches are essential given the proliferation of e-cigarette products and various country-specific regulatory strategies, which can pre-sent challenges for synthesis across individual studies. This Cancer Grand Challenge aims to generate objective, high-quality studies to inform the general public, including adult smokers who may consider using the products to quit smok-ing, clinicians, and public health authorities.

normal PhEnotyPEsIn 1975, Mintz and Illmensee reported establishing a nor-

mal, fertile mouse from a malignant teratocarcinoma line, providing compelling evidence that cancer cells could adopt a normal phenotype (13). More recent studies have demon-strated how sun-exposed skin contains a patchwork of evolving clones, with more than a quarter of the cells containing cancer-associated mutations; yet, despite this, sun-exposed skin main-tains the physiologic functions of normal epidermis (14). This discovery was paradigm shifting, challenging the assumption that cancer driver mutations occurred rarely in long-lived cells and that most existed in malignant tissues too small to be detected in the clinic. Multiple studies from a number of different groups have since corroborated the findings in other tissues, including esophagus, lung, liver, and breast.

We have also known for some time that cancer is associated with the disruption of normal tissue architecture, breakdown of tissue boundaries, stromal changes, and angiogenesis. Early studies demonstrated an integral role for the extracellu-lar matrix (ECM) and stromal microenvironment, including importance of the tensegrity, mechanochemical regulation, in carcinogenesis (15). In addition, early insights from the Cancer Grand Challenges Mutographs team (supported in an earlier round of the initiative) are starting to revive the pro-moter hypothesis, which posits that processes such as wound healing and inflammation may act to promote malignancy in cells harboring oncogenic mutations (16).

With this Cancer Grand Challenge, we ask teams to con-sider what we now know about mutational load in normal cells, combined with cell-extrinsic factors such as the role of the ECM, tissue architecture, metabolite availability, inflam-mation, and the potential role of systemic responses (e.g., the immune system), to uncover the underlying mechanisms that allow these cells to be phenotypically “normal,” and the necessary molecular events that drive them to malignancy.

InFlammatIonWe know there are many environmental and lifestyle fac-

tors that increase an individual’s risk of cancer, but for many risk factors the biological mechanisms by which they lead to cancer development remain elusive. Inflammation is a major biological process that often ensues with exposure to carcino-gens, yet we know relatively little about the cellular and molec-ular mechanisms linking inflammation and carcinogenesis.

The link between inflammation and cancer is likely to be vast and complicated. Dvorak described tumors as wounds that never heal (17), with more recent studies linking the pathology of some tumors with inflammatory processes that occur as a result of normal wound healing (18). In 1941, Rous and Kidd suggested that tumors arose from an initiation state, where cells containing mutations remain normal until a secondary event, such as inflammation, occurs to promote growth of these cells—linking this challenge intimately with elements of the “Normal Phenotype” challenge concept (19) described above.

Many unanswered questions remain about the array of events that can stimulate tumor-enhancing inflammation. How many “types” of inflammation are there and do they each have similar effects on tumor development? What is the

Research. on July 13, 2021. © 2020 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657

Page 3: Cancer Grand Challenges: Embarking on a New Era of Discovery · 2020. 12. 8. · VIEWS January 2021 CANCER DISCOVERY | OF1 In Focus Cancer Grand Challenges: Embarking on a New Era

views

January 2021 CANCER DISCOVERY | OF3

effect of inhibiting inflammation at different stages of tumor development? How does age affect the quality and potency of the immune response?

Through this Cancer Grand Challenge, we hope that teams will examine the causal mechanisms linking inflammation and cancer initiation, to provide opportunities for cancer preven-tion and important markers for diagnosing early-stage cancer.

solId tumors In chIldrEnWith the introduction of chemotherapy in the 1960s and

the use of combination approaches, survival from some pedi-atric cancers has increased dramatically.

This is particularly true for pediatric hematologic cancers. Genomic profiling revolutionized our understanding of the genetic basis of acute lymphoblastic leukemia (ALL), and the use of tyrosine kinase inhibitors with chemotherapy in BCR–ABL-positive ALL is an excellent example of targeted therapy, yielding a 3-year event-free survival of 80%, more than twice that of historical controls (20). Considerable efforts have also led to the development of chimeric antigen receptor (CAR) T-cell therapy for use in pre–B-cell ALL by targeting CD19 (21). CAR T-cell therapy was FDA-approved for B-cell malig-nancies in 2017, and its application continues to expand, particularly for hematologic malignancies.

In general, however, the progress made in pediatric hema-tologic cancers has not been mirrored in solid tumors—with survival rates for some tumor types remaining depressingly low. There are a multitude of reasons behind this lack of progress. First, many of these tumors are rare, mak-ing building research programs difficult. A further criti-cal issue is a lack of effective therapies for pediatric solid tumors, because many new therapies are developed to exploit vulnerabilities present in adult but not pediatric cancers. For treatments that do show efficacy, many result in serious long-term morbidities and associated health complications, including learning difficulties, abnormal growth, and infertility. An additional challenge for brain and central nervous system tumors is the low permeability of the blood–brain barrier.

We know that many pediatric solid tumors are distinct from adult cancers with respect to their biology and how they respond to therapy. We also know that many of these tumors mimic or hijack developmental processes or derive from cells present during development. A deeper under-standing of these features is important if we are to make real progress for pediatric patients. Translating the success of immunotherapies in adult cancers to pediatric solid tumors could be another important opportunity to explore. A global collaborative approach is needed to tackle this Cancer Grand Challenge, advance our understanding of pediatric solid tumors, and translate what we learn into real clinical benefit.

macromolEculEsMacromolecules hold considerable promise as effective

therapeutics, not just to treat cancer but for many other dis-eases. Their structure offers high specificity and potency, and, in comparison with small molecules, often they have lower

nonspecific binding, less toxicity, and reduced drug–drug interactions.

Yet, their potential efficacy is compromised; many bar-riers limit the systemic delivery of macromolecules. Their rapid clearance from the blood hampers biodistribution, and we lack an efficient, safe, and specific delivery mech-anism. Further reducing their therapeutic potential are phagocytic clearance, their large molecular weight affecting biodistribution, their chemical stability, challenges with their solubility, and, critically, their failure to permeate cell membranes.

Efforts are ongoing to overcome these barriers, with a major focus on nanotechnology-based delivery systems, including efforts to exploit receptors on the blood–brain bar-rier. Recently, the use of extracellular vesicles (EV) to deliver cargo such as RNAi, CRISPR/Cas9, and chemotherapeutics to target tissues has shown promise, although efficient EV loading, tissue targeting, and the functional delivery of the therapeutic agents must be further explored (22).

Despite significant advances in the field of macromolecule delivery, an ongoing obstacle remains their inability to cross the cell membrane to enter the cytoplasm and nucleoplasm where target molecules reside. Many molecules that bind to the cell surface enter endosomes but cannot escape that membrane compartment.

With this Cancer Grand Challenge, we invite scientists to develop approaches to overcome this barrier, to achieve the noninvasive, systemic delivery of macromolecules, and to cre-ate new ways to treat cancer.

cachExIaCachexia is a wasting syndrome frequently associated with

cancer and other chronic diseases where sufferers lose weight and experience decline of their overall health. Clinically, cancer cachexia involves dysfunction of multiple tissues and organ systems, drastically decreasing a patient’s quality of life and their tolerance to therapeutics, and is a significant deter-minate in patient survival.

Despite cachexia having such significant clinical impli-cations, we still know relatively little about the condition. Clinical studies have revealed it is not simply a result of nutri-tional deficiency, as nutritional supplements do not reverse cachexia in patients with cancer. In addition, although there are numerous clinical trials of agents targeting mediators of cachexia, there are no effective therapies.

We are, however, starting to learn more about its biol-ogy, recognizing that although muscular atrophy is the major manifestation of the condition, cachexia is a systemic paraneo plastic phenomenon, affecting or influenced by mul-tiple tissues. Furthermore, recent advances have revealed more about muscle atrophy/hypertrophy and adipocyte wasting/browning (23), and the involvement of metabolism and the immune, endocrine, and central nervous systems in cachexia (24).

To develop effective treatment strategies, we need to fully understand the basic mechanisms that underpin this mul-tifactorial condition. This Cancer Grand Challenge invites teams to build on recent advances and provide a platform for the development of therapeutic approaches to reverse this

Research. on July 13, 2021. © 2020 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657

Page 4: Cancer Grand Challenges: Embarking on a New Era of Discovery · 2020. 12. 8. · VIEWS January 2021 CANCER DISCOVERY | OF1 In Focus Cancer Grand Challenges: Embarking on a New Era

Views

OF4 | CANCER DISCOVERY January 2021 AACRJournals.org

debilitating condition, which would significantly improve the lives of patients with cachexia.

dormancyThe issue of dormancy and recurrence following seemingly

effective treatment remains a significant problem in cancer. Some cancers, for example certain types of breast cancer, undergo a period of dormancy, ranging from years to dec-ades, before metastatic disease recurs.

Observations by Willis (1934) and Hadfield (1954) laid the foundations for the field of dormancy, with Willis noting late metastases in patients without postmortem evidence of local recurrence, and Hadfield proposing that recurrence occurs because dormant cancer cells enter a state of temporary mitotic arrest. For decades, metastasis was thought to occur at the end of a process that led to lethal cancer; in 2003, Schmidt-Kittler and colleagues demonstrated that breast cancer cells could disseminate with fewer genomic alterations than previously thought, lying dormant for many years before acquiring the genomic abnormalities that make them metastatic (25).

Elucidating the mechanisms governing dormancy has been challenging for many reasons. Dormancy comprises a num-ber of different states, dependent on the cell type, cell context, oncogenic signaling, availability of nutrients, oxygen levels, growth factors, the ECM, and systemic responses (26). There may, then, be many different ways to induce dormancy, and this is regulated by active mechanisms. Another important barrier to overcome is the development of reliable model systems to study dormancy; mouse or culture models, for example, may not accurately recapitulate recurrence after several decades, as seen in patients.

Insight into the dynamic cross-talk between cancer and the host is of critical importance to uncover the mechanisms that establish long-term dormancy after seemingly success-ful treatment and the mechanisms involved in dormant cell reawakening. This Cancer Grand Challenge seeks to develop and apply innovative approaches to accurately study dormant tumor cells, which could change the way we think about cancer progression. The ability to eliminate dormant tumor cells or prevent their reactivation would have huge therapeutic potential to limit the rates of cancer recurrence.

conclusIonMaking progress in these fields and attempting to solve

these challenges, we believe, demands a new approach. We are seeking to harness ideas from diverse scientific disciplines and the broadest range of experts; with this round of chal-lenges, up to four global, multidisciplinary teams will each receive up to £20m (~$25m) to come together, think differ-ently, and strive for breakthroughs.

From now until April 22, 2021, we invite you to assem-ble your Cancer Grand Challenges team and submit applica-tions (https://cancergrandchallenges.org/researchers) with the potential to unlock new thinking in pursuit of some of the most complex challenges in cancer research.

Authors’ DisclosuresNo disclosures were reported.

AcknowledgmentsThe authors gratefully acknowledge Gemma Balmer-Kemp and

Emily Farthing for support when writing this article.

Published first December 8, 2020.

REfERENCES 1. Alt FW, Kellems RE, Bertino JR, Schimke RT. Selective multiplication

of dihydrofolate reductase genes in methotrexate-resistant variants of cultured murine cells. J Biol Chem 1978;253:1357–70.

2. Alitalo K, Schwab M, Lin CC, Varmus HE, Bishop JM. Homogene-ously staining chromosomal regions contain amplified copies of an abundantly expressed cellular oncogene (c-myc) in malignant neu-roendocrine cells from a human colon carcinoma. Proc Natl Acad Sci 1983;80:1707–11.

3. Turner KM, Deshpande V, Beyter D, Koga T, Rusert J, Lee C, et  al. Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity. Nature 2017;245:122–5.

4. Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res 1965;37:614–36.

5. Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BC, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011;479:232–6.

6. Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell 2014;31:722–33.

7. Wang C, Vegna S, Jin H, Benedict B, Lieftink C, Ramirez C, et  al. Inducing and exploiting vulnerabilities for the treatment of liver cancer. Nature 2019;574:268–72.

8. Malas M, van der Tempel J, Schwartz R, Minichiello A, Lightfoot C, Noormohamed A, et al. Electronic cigarettes for smoking cessation: a systematic review. Nicotine Tob Res 2016;18:1926–36.

9. Hartmann-Boyce J, McRobbie H, Lindson N, Bullen C, Begh R, Theodoulou A, et  al. Electronic cigarettes for smoking cessation. Cochrane Database Syst Rev 2020;10.

10. National Academies of Sciences, Engineering, and Medicine. Public health consequences of E-cigarettes. Washington, DC: The National Academies Press; 2018.

11. Eltorai AEM, Choi AR, Eltorai AS. Impact of electronic cigarettes on various organ systems. Respir Care 2019;64:328–36.

12. National Center for Chronic Disease Prevention and Health Promo-tion (US) Office on Smoking and Health. E-cigarette use among youth and young adults. Atlanta, GA: Centers for Disease Control and Prevention; 2016.

13. Mintz B, Illmensee K. Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc Natl Acad Sci 1975;72:3585–9.

14. Martincorena I, Roshan A, Gerstung M, Ellis P, Van Loo P, McLaren S, et al. High burden and pervasive positive selection of somatic muta-tions in normal human skin. Science 2015;348:880–6.

15. Ingber DE. Cancer as a disease of epithelial–mesenchymal interac-tions and extracellular matrix regulation. Differentiation 2002;70: 547–60.

16. Riva L, Pandiri AR, Yun RL, Droop A, Hewinson J, Quail MA, et al. The mutational signature profile of known and suspected human carcinogens in mice. Nat Genet 2020;54:1189–97.

17. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 1986;315: 1650–9.

18. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420: 860–7.

19. Rous P, Kidd JG. Conditional neoplasms and subthreshold neoplas-tic states: a study of the tar tumors of rabbits. J Exp Med 1941;73: 365–90.

20. Schultz KR, Bowman PW, Aledo A, Slayton WB, Sather H, Devidas M, et al. Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a children’s oncology group study. J Clin Oncol 2009;27:5175–81.

Research. on July 13, 2021. © 2020 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657

Page 5: Cancer Grand Challenges: Embarking on a New Era of Discovery · 2020. 12. 8. · VIEWS January 2021 CANCER DISCOVERY | OF1 In Focus Cancer Grand Challenges: Embarking on a New Era

views

January 2021 CANCER DISCOVERY | OF5

21. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter D, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368:1509–18.

22. Melling GE, Carollo E, Conlon R, Simpson JC, Carter DRF. The challenges and possibilities of extracellular vesicles as therapeutic vehicles. Eur J Pharm Biopharm 2019;144:50–6.

23. Vegiopoulos A, Rohm M, Herzig S. Adipose tissue: between the extremes. EMBO J 2017;36:1999–2017.

24. Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers 2018;4:17105.

25. Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M, Ahr A, Blank-enstein TJF, et al. From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression. Proc Natl Acad Sci U S A 2003;100:7737–42.

26. Endo H, Inoue M. Dormancy in cancer. Cancer Sci 2019;110: 474–80.

Research. on July 13, 2021. © 2020 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657

Page 6: Cancer Grand Challenges: Embarking on a New Era of Discovery · 2020. 12. 8. · VIEWS January 2021 CANCER DISCOVERY | OF1 In Focus Cancer Grand Challenges: Embarking on a New Era

Published OnlineFirst December 8, 2020.Cancer Discov   Iain Foulkes and Norman E. Sharpless  DiscoveryCancer Grand Challenges: Embarking on a New Era of

  Updated version

  10.1158/2159-8290.CD-20-1657doi:

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerdiscovery.aacrjournals.org/content/early/2020/12/08/2159-8290.CD-20-1657To request permission to re-use all or part of this article, use this link

Research. on July 13, 2021. © 2020 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Published OnlineFirst December 8, 2020; DOI: 10.1158/2159-8290.CD-20-1657