VOLUME

32



NUMBER

15



MAY

20

2014

JOURNAL OF CLINICAL ONCOLOGY

A S C O 50TH A N N I V E R S A R Y

Progress Against GI Cancer During the American Society of Clinical Oncology’s First 50 Years Robert J. Mayer, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA Alan P. Venook, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA Richard L. Schilsky, American Society of Clinical Oncology, Alexandria, VA

Tumors of the GI tract were frequent causes of cancer-related mortality in 1964 when the American Society of Clinical Oncology (ASCO) was founded and remain so in 2014, 50 years later. GI cancers accounted for 31% of the reported 305,000 cancer-related deaths in the United States in 19671 and 28% of the anticipated 585,720 such deaths in 2014.2 Worldwide, GI cancers led to more than 2.3 million deaths in 2008, far more than deaths resulting from any other group of malignant conditions.3 In 1964, studies related to GI cancer were in their infancy. Newly formed cooperative groups were focused on clinical trials dealing with hematologic malignancies. The chemotherapeutic agent that remains the cornerstone of treatment for GI cancer (fluorouracil [FU]) had recently been synthesized and was being used in early clinical trials at the Mayo Clinic and the University of Wisconsin. The combination of FU and radiation therapy was being explored in the management of locally advanced rectal cancer.4 In 1973, the National Cancer Institute organized the GI Tumor Study Group (GITSG), a multidisciplinary clinical research collaborative from which seminal clinical trials emerged over the subsequent 15 years. Substantial progress in GI oncology has occurred since then, much of which has been specific to individual diseases but also as a result of advances in oncology in general. The creation of the SEER program of the National Cancer Institute and the Nurses’ Health Study and the Health Professionals Follow-Up Study of the Harvard School of Public Health has provided population-based cohorts from which have emerged valuable epidemiologic information, leading to a better understanding of both genetic and environmental risk factors (eg, smoking, obesity, viral carcinogenesis) for cancer. Technological advances such as computed tomography, magnetic resonance imaging, positron emission tomography, fiberoptic endoscopy, endoluminal stents, and the development of immune and molecular biomarkers to refine prognostic and predictive information have all contributed to improved outcomes for patients with GI cancers. Surgical morbidity and outcomes have improved as a result of refinements in technique (such as better-tolerated Whipple resections and the introduction of total mesorectal excisions and minimally invasive resections of the esophagus, stomach, and colorectum), and the introduction of linear accelerators has reduced injury to normal tissues after radiation therapy. The development of effective cytotoxic and targeted drugs in addition to FU and the validation of Journal of Clinical Oncology, Vol 32, No 15 (May 20), 2014: pp 1521-1530

Table 1. Changing 5-Year Survival Expectations in the United States for GI Cancers (1975-1977 v 2003-2009)2 5-Year Survival (%) Cancer Site

1975-1977

2003-2009

Esophagus Gastric Pancreas Hepatobiliary Colon/rectum

5 15 2 3 50

19 29 6 18 66

adjuvant and neoadjuvant treatment strategies have improved clinical outcomes, particularly for patients with colorectal cancer2 (Table 1). Advances in specific GI cancers are presented in subsequent sections of this article. Esophageal Cancer In 1964, the vast majority of esophageal cancers occurring in the United States were squamous cell carcinomas arising in the cervical and thoracic esophagus,5 most frequently in older males with a history of excess alcohol and/or tobacco use.6 Squamous cell carcinomas are still the most frequently observed form of esophageal cancer worldwide, where the disease is the sixth leading cause of cancer-related mortality.3 The incidence of squamous cell esophageal cancer has decreased in the United States during the past 50 years, whereas the incidence of adenocarcinoma of the esophagus has increased sevenfold, such that adenocarcinoma now comprises more than 75% of esophageal tumors.7 These adenocarcinomas, which occur far more often in men than women, arise in the distal esophagus in the presence of chronic gastric reflux,8 often in association with Barrett’s esophagus.9 Chronic gastric reflux, Barrett’s esophagus, and esophageal adenocarcinomas occur more commonly in obese individuals.10 These tumors arise within the dysplastic columnar epithelium of the distal esophagus with aneuploidy and mutations of the p53 gene detectable in the dysplastic epithelium before progression to frank neoplasia.11 Fifty years ago, surgery was the mainstay of treatment for patients with esophageal cancer. Operative procedures were poorly tolerated,12 in part because patients were suboptimal surgical candidates because of malnutrition and the frequent presence of significant comorbid © 2014 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

1521

Mayer, Venook, and Schilsky

Table 2. Important Observations and Advances in Esophageal Cancer Between 1964 and 2014 Observation/Advance Marked increase in adenocarcinomas in the United States Linkage of the development of adenocarcinomas to obesity, chronic gastric reflux, and Barrett’s esophagus Preoperative chemoradiation therapy extends survival PET scans provide more accurate staging and offer rapid early assessment of responsiveness to treatment Abbreviation: PET, positron emission tomography.

illness. The probability of 5-year survival in the mid-1970s for American patients with esophageal cancer was approximately 5% (Table 1). In more recent years, surgery has been better tolerated in patients with distal esophageal tumors; the development of minimally invasive surgical techniques has further reduced perioperative morbidity.13 Although treatment outcomes are slightly improved if preoperative radiation therapy or platinum-based chemotherapy is administered, the best survival outcomes are achieved using preoperative chemotherapy combined with radiation therapy14,15 with some reports that focus on squamous cell cancers suggesting no additional benefit when surgery is added if significant shrinkage of tumor has been achieved through the use of chemoradiotherapy.16,17 Sequential positron emission tomography scans seem to be useful in assessing responsiveness to neoadjuvant therapy.18 At present, the estimated likelihood of 5-year survival for American patients is 19%, which still indicates that long-term benefit can be achieved in only a disappointing minority of patients while also demonstrating a fourfold improvement over the 5% likelihood of 5-year survival of 30 years ago2 (Table 1). Major observations and advances in esophageal cancer during the past 50 years are summarized in Table 2. Gastric Cancer The dramatic and poorly explained decrease in the incidence and mortality rates of gastric cancer in the United States that began in 1930 has continued in the past 50 years. Whereas gastric cancer was the cause of 5.7% of cancer-related deaths in the United States in 1967,1 it will result in only 1.9% of the anticipated cancer-related deaths in 2014.2 Nonetheless, gastric cancer remains the second most frequent cause of cancer mortality worldwide.3 The risk for gastric cancer is greater among lower socioeconomic populations.19 Migrants from high- to low-incidence regions maintain their susceptibility to gastric cancer, whereas the risk for their offspring approximates that of the prevailing risk of their new homeland.20 These observations suggest that an environmental exposure, probably beginning early in life, is related to the development of gastric cancer, with dietary carcinogens21 and Helicobacter pylori22 infection being considered contributory factors. During the past 50 years, gastric adenocarcinomas have been subdivided into two types23: an intestinal type characterized by ulcerative lesions in the antrum and lesser curvature preceded by atrophic gastritis and often initiated by H pylori infection; and a diffuse type, which occurs throughout the stomach, often without mucosal ulceration, resulting in a lack of distensibility of the gastric wall (linitis plastica), associated with the loss of expression of E-cadherin and carrying a worse prognosis. Although the incidence of diffuse carcino1522

© 2014 by American Society of Clinical Oncology

mas is similar in most populations, the intestinal type tends to predominate in high-risk geographic regions and is less likely to be found in areas where the frequency of gastric cancer has been declining. A germline mutation in the E-cadherin gene (CDH1) inherited in an autosomal dominant pattern and coding for a cell adhesion protein has been associated with a high incidence of occult diffuse-type gastric cancer in young, asymptomatic carriers.24 During the past two decades, the use of double-contrast radiographic examinations has been replaced by computed tomography scanning and esophagogastroscopy for the evaluation of individuals with epigastric complaints. Although a subtotal gastrectomy remains the operative procedure of choice for patients with distal, intestinaltype carcinomas,25 many patients with diffuse carcinomas now receive preoperative chemotherapy or chemoradiotherapy given that surgical outcomes for these individuals have generally been poor. In highincidence regions of the world such as Asia, large numbers of lymph nodes are routinely removed as part of potentially curative operations for gastric cancer. The value of these so-called D2 and D3 procedures has been assessed in several randomized trials in comparison with resections that remove fewer lymph nodes. These studies have shown that the incremental benefit of the more aggressive surgery is quite modest, possibly because some of the benefit in tumor control achieved by the D2/D3 procedure is offset by increased perioperative morbidity and mortality.26 Several FU-based chemotherapy combinations such as FU/leucovorin/oxaliplatin (FOLFOX), epirubicin/cisplatin/FU (ECF), and docetaxel/cisplatin/FU (TCF) have been studied as potential treatment for patients with advanced gastric cancer. Although response rates vary, the median survival time is generally less than 12 months, with the regimens differing primarily in the severity of adverse effects and cost.27 The addition of trastuzumab to chemotherapy in the 15% of patients with human epidermal growth factor receptor 2/neu overexpressing tumors extends the median survival by 2.7 months.28 Adding bevacizumab to chemotherapy has not prolonged median survival.29 However, preliminary experience using a more recently developed anti–vascular endothelial growth factor receptor monoclonal antibody, ramucirumab, seems promising.30,31 Adjuvant or perioperative chemotherapy has been shown to enhance surgical cure in studies conducted in Asia32,33 and Europe.34 In the United States, perhaps because of the use of somewhat less extensive surgical procedures, the best outcomes have been achieved with chemoradiotherapy after surgery,35 although no added benefit was found when a more intensive chemotherapy regimen (ECF) was compared with FU and leucovorin alone as part of an adjuvant chemoradiotherapy program.36 During the last 30 years, the likelihood of 5-year survival for American patients with gastric adenocarcinoma has increased from 15% to 29%2 (Table 1). Major observations and advances in gastric cancer during the past 50 years are summarized in Table 3. Pancreatic Cancer In 1964, little was known about pancreatic cancer; its cause was unknown, the pancreas could not be adequately imaged, and the disease was almost always far advanced when discovered. On the rare occasions when the diagnosis could be made early, the only treatment was the Whipple procedure, a complex operation associated at that time with high rates of perioperative morbidity and even mortality and which was rarely curative.37 JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

ASCO 50th Anniversary

Table 3. Important Observations and Advances in Gastric Cancer Between 1964 and 2014 Observation/Advance Incidence and mortality rates in the United States continue to decrease Gastric cancer occurs in two subtypes: Intestinal (ulcerated lesions in antrum after atrophic gastritis) Diffuse (submucosal infiltration causing lack of distensibility) Helicobacter pylori infection is associated with the intestinal subtype A germline mutation in the CDH1 gene, which regulates E-cadherin, is associated with familial gastric cancer The small survival benefit achieved with extended (D2 and D3) surgical resections may be offset by increased perioperative morbidity and mortality Adjuvant chemoradiation therapy prolongs survival

Although pancreatic cancer remains a sinister disease today and represents the fourth most common cause of cancer-related death in the United States, small improvements in outcome are becoming evident. More sophisticated imaging has made the pancreas visible to clinicians, and advances such as endoscopic retrograde cholangiopancreatography38 and transduodenal fine-needle biopsy via an endoscope39 can lead to an earlier diagnosis. Additionally, surgical techniques and postoperative care have improved, with patients being referred to high-volume centers, leading to a marked reduction in postoperative morbidity and mortality.40 Problems such as jaundice, pain, weight loss, anorexia, and hypercoagulability can still be overwhelming early in the course of pancreatic cancer, but aggressive symptom management and the appropriate use of endoluminal stents to relieve biliary tree or gastric outlet obstruction have led to better palliation for most patients. A hallmark of pancreatic cancer is the dense fibrous stroma that often envelops the tumor; this sclerotic process limits the accessibility of tumor tissue, accounting in part for the slow pace of progress in understanding the molecular basis of the disease.41 It is now known that RAS mutations are present in more than 90% of cancers,42 but other key molecular drivers have not yet been identified. The CA19-9 is an accepted serologic marker43 in the disease and is of some clinical use in monitoring the course of the illness. Recently, a transgenic mouse model that spontaneously recapitulates the stroma-laden human disease44 has been introduced and may facilitate the more rapid development of biologic insights regarding the initiation and progression of pancreatic cancer. Epidemiologic studies have linked the development of pancreatic to obesity,45 the recent onset of type II diabetes,46 and cigarette smoking.47 Investigations in families at higher-than-usual-risk for pancreatic cancer have revealed an increased incidence in the setting of Peutz-Jegher syndrome,48 hereditary nonpolyposis colon cancer,49 and mutations in the BRCA1 or, particularly, the BRCA2 genes.50 Sequencing studies have revealed putative pancreatic cancer susceptibility genes,51 but these observations have not yet altered clinical practice or identified individuals in whom intensive screening procedures might be beneficial. Between 1985, when the GITSG established FUchemoradiotherapy as standard adjuvant therapy,52 and 1997, when modest benefits in survival and symptom control led to US Food and Drug Administration approval of gemcitabine,53 the lack of therapeutic progress was disheartening. During the ensuing years, gemcitabine www.jco.org

has been shown to be effective in the adjuvant setting,54 but the role of radiation therapy as part of adjuvant treatment has been questioned.55 It is sobering to note that the median survival time of 20 to 24 months in contemporary adjuvant studies54-57 is no better today than it was in the GITSG report of 1985.52 Given the availability of improved staging techniques, this observation suggests that whatever progress has been made with presently available treatment approaches has been minimal and has not significantly impacted the natural history of the disease. Between 1997 and 2011, despite a host of attempts to combine gemcitabine with other drugs, no significant improvements were made in the treatment of advanced pancreatic cancer.58 Since 2011, the introduction of the FOLFIRINOX (fluorouracil, leucovorin, irinotecan, oxaliplatin) and gemcitabine/NAB-paclitaxel regimens seems to have changed the dynamic. The use of FOLFIRINOX has more than tripled the response rate compared with gemcitabine and has extended the median survival from 5 months to more than 11 months59 while maintaining an acceptable quality of life.60 The administration of the gemcitabine/NAB-paclitaxel doublet has improved the median overall survival achieved with gemcitabine from 5 months to 8.5 months.61 Whether one of these chemotherapy regimens is superior to the other and whether either might prove to be effective in the adjuvant setting are not yet known, but both give hope that the rate of progress over the next 5 years will vastly exceed that of the past 50 years. During the past 30 years, the probability of 5-year survival for American patients with pancreatic cancer has increased slightly from 2% to 6%2(Table 1). Major observations and advances in pancreatic cancer during the past 50 years are summarized in Table 4. Hepatobiliary Cancers In 1964, hepatocellular carcinoma was thought to be primarily a consequence of alcoholic cirrhosis. In large part as a result of the efforts of Beasley et al,62 it is now clear that alcohol accounts for just a small fraction of the global burden of hepatocellular carcinoma with the majority of cases resulting from chronic infection with hepatitis B or hepatitis C virus. Through the prospective surveillance of male governmental employees in Taiwan, the risk for developing hepatocellular carcinoma was shown to be increased by 390-fold in individuals with serologic evidence of hepatitis B infection. The subsequent development of a vaccine against hepatitis B and its administration to

Table 4. Important Observations and Advances in Pancreatic Cancer Between 1964 and 2014 Observation/Advance The development of a transgenic mouse model that recapitulates the human disease Epidemiologic link to obesity, recent onset of type II diabetes, and chronic cigarette smoking Increased incidence in families with a history of Peutz-Jegher syndrome, hereditary nonpolyposis colon cancer, and BRCA syndromes Introduction of endoscopic retrograde cholangiopancreatography, allowing the placement of stents to nonsurgically relieve tumor-related obstructive jaundice Improved surgical technique leading to decreased operative morbidity and mortality Successful use of gemcitabine as adjuvant therapy and FOLFIRINOX and gemcitabine/NAB-paclitaxel to treat metastatic disease Abbreviation: FOLFIRINOX, fluorouracil, leucovorin, irinotecan, oxaliplatin.

© 2014 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

1523

Mayer, Venook, and Schilsky

Taiwanese neonates born to mothers who were hepatitis B carriers essentially eradicated hepatocellular carcinoma in a generation of Taiwanese citizens,63 confirming the causative relationship between hepatocellular carcinoma and hepatitis B. The present inability to develop a vaccine against hepatitis C has resulted in continued high rates of hepatocellular carcinoma infection in endemic areas such as Africa but also in the United States and Europe.64 The development of orthotopic liver transplantation during the past 50 years has provided a dramatic and potentially curative form of therapy for some patients with hepatocellular carcinoma. The process of organ allocation for such patients remains controversial, but experience gathered during the 1990s in selected patients has confirmed the feasibility and long-term benefit of liver transplantation in patients with hepatocellular carcinoma.65 The original eligibility criteria for transplantation—three or fewer hepatic nodules and the absence of major blood vessel involvement and extrahepatic tumor spread— have subsequently been liberalized66 to the extent that approximately 17% of all liver transplantation procedures performed in the United States are now carried out on patients with hepatocellular carcinoma.67 For patients with hepatocellular carcinoma not suitable for liver transplantation, standard cytotoxic chemotherapy has produced disappointing results. Therapeutic efforts have focused on a unique feature of such hepatic tumors: their hypervascularity. The use of transhepatic arterial chemoembolization has proven to be effective in providing transient local tumor control and even an apparent slight prolongation in survival.68 However, hepatocellular carcinoma ultimately represents a systemic rather than a local disease; the poor hepatic reserve of patients with hepatocellular carcinoma and their frequent comorbid conditions have hampered the development of effective systemic treatment. Sorafenib, a tyrosine kinase inhibitor thought to target the RAF gene, was the first drug approved for the treatment of hepatocellular carcinoma on the basis of studies conducted in Asia69 and the United States and Europe.70 These pivotal trial results only emerged after it was decided to limit entry to patients with adequate liver function, thereby minimizing the impact of an impaired liver on the outcome of the study and offering a more meaningful setting to better examine the efficacy of the study drug.71 Biliary tract cancers (ie, cholangiocarcinoma, gall bladder cancer, and ampullary cancer) have also proven to be difficult to study and to treat. Although once considered uncommon, the introduction of cytokeratin immunohistochemical staining and the availability of circulating tumor markers such as CA19-9 have indicated that they occur more commonly than previously recognized. The observation in 2010 that the combination of gemcitabine and cisplatin was superior to gemcitabine alone72 has led to this regimen becoming the standard treatment against which new combinations are being compared. The probability of 5-year survival for American patients with hepatobiliary cancer has increased from 3% to 18% during the past 30 years2 (Table 1). Major observations and advances in hepatobiliary cancers during the past 50 years are summarized in Table 5. Pancreatic Neuroendocrine and Intestinal Carcinoid Tumors In contrast to the outlook for the majority of GI cancers, the prognosis for patients with GI neuroendocrine cancers has signifi1524

© 2014 by American Society of Clinical Oncology

Table 5. Important Observations and Advances in Hepatocellular Carcinoma Between 1964 and 2014 Observation/Advance Chronic infection with hepatitis B or hepatitis C virus is the most common causative factor and the incidence of hepatocellular carcinoma can be markedly reduced by vaccination against hepatitis B Curative potential of orthotopic liver transplantation in highly selected patients Palliative benefit of transhepatic chemoembolization Sorafenib prolongs survival in patients with preserved liver function

cantly improved during the past 50 years. The discovery of somatostatin in 197373 led to a series of advances that have transformed the pancreatic neuroendocrine tumors (PNET) and intestinal carcinoid tumors into manageable, chronic diseases. Because both of these entities can have clinical syndromes that result from hormone excess, they have often been considered to be a single pathologic entity. The introduction of octreotide (an octapeptide analog of somatostatin with a longer half-life than the native molecule that is capable of blocking hormone release)74 has been shown to diminish the associated symptoms experienced by patients with both carcinoid tumors (eg, overwhelming diarrhea) and PNETs (eg, abnormal glucose metabolism from the hormonal effects of insulin or glucagon). The widespread use of abdominal imaging along with the ability to biopsy lesions deep within the abdomen may account for the apparent increasing frequency of these diagnoses. It is now clear that most PNETs do not secrete hormones and that the cellular proliferative index may be the best predictor of prognosis. Advances such as somatostatin and its sustained-release formulations allow many of these patients to live longer with a better quality of life.75 Patients with carcinoid disease may live for decades with a chronic condition, whereas patients with PNETs typically succumb to cancer progression after approximately 5 to 8 years. During the last several years, the ability to target metabolic and angiogenesis pathways with oral tyrosine kinase inhibitors76 and with everolimus77 has provided new treatment options and stimulated new research approaches for these rare diseases. Colorectal Cancer At the birth of ASCO in 1964, little was known about the causes of colorectal cancer; treatment was largely limited to surgical resection of the primary tumor, and FU was used with marginal effectiveness in treating patients with metastatic disease. Since then, substantial progress has been made in the early detection, prevention, and treatment of colorectal cancer leading to significant reductions in the incidence and mortality in the United States and most resource-rich countries.2 Observational studies have linked the consumption of red meat,78 obesity,79 and lack of physical activity80 to an increased risk of colon cancer; the regular use of aspirin seems to reduce that risk.81,82 Aspirin use83 and vigorous physical activity84 also seem to decrease the risk of recurrence and death among patients already diagnosed with colon cancer. Familial colon cancer syndromes such as familial adenomatous polyposis and hereditary nonpolyposis colon cancer have been well characterized both clinically and genetically, and the introduction of genetic counseling services for high-risk families has enabled comprehensive risk assessment and early intervention to reduce JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

ASCO 50th Anniversary

the risk of colorectal cancer in affected individuals.85 Sporadic colonic adenomas are now generally accepted to be premalignant lesions with the risk of malignancy related to polyp size, villous morphology, and sessile appearance.86 Thus, screening the general population for colorectal polyps and cancer, previously with fecal occult blood testing and more recently by direct visualization through sigmoidoscopy or colonoscopy, has become established and has been proven to reduce cancer incidence and mortality.87-89 Unfortunately, the proportion of the eligible population who actually undergo screening remains suboptimal, largely as a result of misunderstandings regarding individual risk, poor access to care, and concerns about the discomfort of the bowel preparation before endoscopy. In recent years, new technologies, such as computed tomographic colonography, have emerged as effective screening strategies, at least for larger polyps, and may be more acceptable to the public than endoscopic procedures; however, the widespread use of computed tomographic colonography will likely be limited by the continued need for bowel preparation, cost, and the availability of radiologists with the necessary expertise to interpret the images.90 Fecal DNA testing for mutations in shed epithelial cells offers the promise of a screening test that is far more specific than testing for fecal occult blood, but this test will not eliminate the need for direct visualization of the colon to identify and remove the source of the abnormal cells.91 Among the most important advances of the last 50 years has been the delineation of the molecular pathways leading to colorectal cancer, described initially by Vogelstein et al92 and Markowitz et al93 and subsequently refined by the Cancer Genome Atlas Network.94 The microsatellite instability pathway, related to mutation or hypermethylation of DNA mismatch repair genes, and the chromosome instability pathway, associated with tumor suppressor gene deletion or oncogene mutation, are now known to lead to aberrations in specific molecular pathways such as WNT, MAPK, PI3K, TGFB, and p53 that contribute to the formation of the majority of colorectal tumors. These new insights will no doubt lead to more effective early detection, prevention, and treatment strategies in coming years.93 The prognosis for newly diagnosed patients with colorectal cancer is primarily related to tumor stage as determined by the TNM classification.95 Other pathologic (eg, tumor perforation, obstruction, perineural and/or vascular invasion, lymphocytic infiltration, number of involved nodes, and total number of nodes in the resected specimen96,97), clinical (eg, elevated preoperative carcinoembryonic antigen level98), and molecular characteristics that may impact prognosis have also been recognized. Among the latter, high microsatellite instability in colon cancer resulting from mutation in certain DNA mismatch repair genes has consistently been associated with more favorable outcomes99 and may predict lack of responsiveness to FU chemotherapy as well.100 BRAF mutations are emerging as an independent adverse prognostic feature,101,102 and several gene expression profiling tests have been introduced that seem useful in refining the prognosis (albeit not presently the management) of patients with stage II disease.103-105 The primary treatment of colon cancer is surgical resection, and several prospective randomized clinical trials have now shown that laparoscopic colectomy produces similar outcomes with reduced morbidity compared with open colectomy.106 Since a series of randomized clinical trials conducted in the 1980s and 1990s clearly established the value of 6 months of postoperative adjuvant chemotherapy for patients with stage III colon cancer, the major subsequent advance www.jco.org

has been the addition of oxaliplatin to FU/leucovorin (ie, FOLFOX).107 Irinotecan was shown to be ineffective when added to FU/ leucovorin in the adjuvant setting.108,109 Surprisingly, neither bevacizumab110 nor cetuximab,111 when added to chemotherapy, has been shown to be useful in the adjuvant setting either, although both drugs are effective in the treatment of patients with metastatic disease. The primary treatment of rectal cancer has evolved considerably in the last 50 years, in large part as a result of information generated from prospective randomized clinical trials conducted primarily outside the United States. Optimal local control and reduced perioperative and long-term morbidity can be achieved with a total mesorectal excision of the rectum after combined chemotherapy (infusional FU or capecitabine) and pelvic radiation and the subsequent administration of adjuvant systemic treatment.112,113 A definitive randomized comparison of administering chemoradiotherapy before or after surgery has demonstrated no significant difference in survival between these two approaches after 11 years of follow-up, but a reduced risk of local recurrence and late complications favors the preoperative treatment.114 In the surgical management of both colon and rectal cancer, outcome is related to clinical volume and the experience of the surgical team with the best outcomes achieved in the high-volume centers.115 The treatment of patients with metastatic colorectal cancer has changed substantially in the last 50 years; although most patients with metastatic disease remain incurable, the median survival has now been prolonged from 6 to 9 months to longer than 2 years. The introduction in the 1970s of the carcinoembryonic antigen as a serum marker, useful in monitoring the course of the disease, facilitated the early detection of disease recurrence after completion of initial treatment. Indeed, a 2006 ASCO clinical practice guideline recommended the measurement of the carcinoembryonic antigen every 3 months for at least 3 years after the completion of initial treatment for patients who are candidates for additional antitumor treatment.116 Up to 25% of patients with a limited number of metastatic sites confined to a single organ (typically liver) may be curable through surgical resection. Other ablative techniques such as radiofrequency ablation, cryoablation, and localized external-beam radiotherapy have shown promise as well, although a 2009 ASCO clinical evidence review called for additional research on radiofrequency ablation before its widespread adoption.117 For the majority of patients with more widely metastatic disease, systemic chemotherapy remains the treatment of choice. Treatment programs in the 1980s focused on optimizing the delivery of FU and enhancing its activity with the addition of leucovorin, a reduced folate that enhances the binding of FU to its target enzyme, thymidylate synthase.118 New cytotoxic drugs for the treatment of colorectal cancer began to be introduced during the late 1990s with the development of irinotecan119 and oxaliplatin,120 both of which improved survival for patients with metastatic colorectal cancer when added to FU and leucovorin, optimally administered in an infusional schedule. These regimens (FOLFIRI and FOLFOX respectively) have been shown in clinical trials to produce similar outcomes, although they differ substantially in their toxicity profiles.121 Capecitabine, an oral prodrug of FU, was also introduced, and in a series of randomized trials in both the metastatic and adjuvant settings, was shown to produce similar outcomes to FU-based chemotherapy.122,123 The next major advance came with the addition of bevacizumab, an anti–vascular endothelial growth factor receptor monoclonal antibody, to cytotoxic chemotherapy as part of the bolus © 2014 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

1525

Mayer, Venook, and Schilsky

FU, leucovorin, irinotecan regimen known as IFL. The registrationdirected clinical trial comparing IFL plus placebo with IFL plus bevacizumab resulted in an improvement in median overall survival from 15.6 to 20.3 months and led to the initial US Food and Drug Administration approval of this first-in-class antiangiogenic treatment.124 Subsequent studies supported its use in combination with both irinotecan- and oxaliplatin-based regimens, and the drug is now widely used as part of first-line chemotherapy treatment for metastatic colorectal cancer.125 Anti– epidermal growth factor receptor (EGFR) monoclonal antibodies also have an established role in the treatment of colorectal cancer.126 First developed and approved as a single-agent treatment for patients with EGFR-expressing metastatic colorectal cancer after progression on standard treatments, cetuximab and panitumumab use is now limited to the treatment of patients with KRAS wild-type tumors (approximately 60% of colorectal cancers).127 The analysis of multiple clinical trial data sets has demonstrated that these drugs are not effective in colorectal tumors that harbor mutations in KRAS and NRAS genes (and likely the BRAF gene) and may even result in inferior outcomes when administered to patients whose tumors have such mutations.128,129 Thus, testing for the presence of mutations in the KRAS gene, and potentially other RAS family genes as well, is now standard practice when deciding whether to prescribe an anti-EGFR antibody for the treatment of patients with metastatic colorectal cancer, either as a single agent or in combination with chemotherapy.130 Although bevacizumab and anti-EGFR antibodies are each effective when combined with chemotherapy in the treatment of metastatic colorectal cancer, several clinical trials have shown that combining thesetargetedagentsproducesinferioroutcomescomparedwithchemotherapy plus bevacizumab alone.131,132 Ongoing studies that directly compare bevacizumab with cetuximab used with chemotherapy as initial treatment for patients with metastatic disease will determine the superior approach; an initial report of a recently completed randomized study suggests that cetuximab may be superior to bevacizumab when added to FOLFIRI in the treatment of patients with metastatic KRAS wild-type colon cancer.133 Two other targeted agents have recently been approved for treatment of advanced colorectal cancer, ziv-aflibercept134 and regorafenib,135 although their modest benefit and high cost has stimulated debate regarding their clinical value in this group of patients. Since 1975, the probability for 5-year survival for American patients with colorectal cancer has increased from 50% to 66%2 (Table 1). Major observations and advances in colorectal cancer during the past 50 years are summarized in Table 6. Anal Cancer In 1964, anal cancer was thought to be an uncommon malignancy, arising most frequently in women with a history of condylomata acuminata (otherwise known as venereal warts).136 Standard treatment at that time was an abdominal-perineal resection, necessitating a permanent colostomy. Despite such an extensive procedure, recurrences developed in more than 25% of patients.137 Much progress has been made against anal cancer during the past 50 years. It is now known that anal cancer is caused by human papillomavirus (HPV) infection transmitted through sexual activity.138 The risk for anal cancer is increased among homosexual males, presumably related to anal intercourse.139 Anal cancer risk is increased in both men and women with AIDS,140 perhaps because their immuno1526

© 2014 by American Society of Clinical Oncology

Table 6. Important Observations and Advances in Colorectal Cancer Between 1964 and 2014 Observation/Advance Elucidation of the molecular evolution of colorectal cancer from normal mucosa to polyp to tumor Several familial colon cancer syndromes described and their genetic underpinnings identified Sporadic adenomatous polyps recognized as premalignant lesions Recognition of the impact of diet, obesity, physical activity, and aspirin use on colon cancer risk Effective screening strategies introduced to decrease incidence and mortality Adjuvant chemotherapy for colon cancer and chemoradiotherapy for rectal cancer prolongs survival and has become the standard of care, particularly for stage III disease Laparoscopic colectomy and total mesorectal excision lead to reduced morbidity and improved outcomes for patients with colon and rectal cancer, respectively New cytotoxic and targeted therapy have led to the improved survival of patients with metastatic colorectal cancer

suppressed state predisposes them to HPV infections. Preliminary data suggest that vaccination against HPV may reduce the likelihood of developing anal cancer.141 In 1974, a colorectal surgeon named Norman Nigro published the initial results of his experience treating patients with anal cancer with several weeks of radiation therapy, infusional FU, and mitomycin.142 This treatment program, which became known as the Nigro regimen, resulted in pathologic complete response in more than 80% of patients whose primary cancers were less than 3 cm in size; with less than 10% of these patients experiencing tumor recurrences, the so-called Nigro regimen has led to cure in approximately 70% of patients without surgery and without the need for a colostomy.143 Randomized trials conducted during the past 20 years have confirmed the advantage of adding chemotherapy (ie, FU/mitomycin) to radiation,144,145 the value of including mitomycin in the chemotherapy regimen,146 and the superiority of FU/mitomycin compared with FU/cisplatin.147 In 2014, 6,260 (86.8%) of the 7,210 individuals anticipated to develop anal cancer are projected to be cured of their disease.2 Major observations and advances in anal cancer during the past 50 years are summarized in Table 7. Summary GI cancers are among the most common tumors and represent the leading cause of cancer-related death worldwide.3 Most of the

Table 7. Important Observations and Advances in Anal Cancer Between 1964 and 2014 Observation/Advance Anal cancer is a sexually transmitted condition associated with HPV infection Increased risk in patients with AIDS The risk for developing anal cancer may be reduced through vaccination for HPV The high colostomy-free cure rate associated with radiation/FU/mitomycin treatment originally described nearly 50 years ago remains the standard of care Abbreviations: FU, fluorouracil; HPV, human papillomavirus.

JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

ASCO 50th Anniversary

progress against these cancers over the last 50 years has resulted from either earlier detection of premalignant lesions or to improved treatment linked to a better understanding of the molecular drivers of malignancy. However, much remains unknown about the fundamental biology of gastric, pancreatic, and biliary tract cancers, and although many of the molecular pathways of colon and rectal cancer have now been elucidated, few effective targeted agents have yet been introduced into clinical practice for these diseases. Although the relatively easy access of the GI tract to endoscopic surveillance and imaging should enable widespread screening of high-risk populations for premalignant and early-stage lesions, much remains to be done to widely disseminate screening programs in many communities. A recent colon cancer screening pilot program in the state of Delaware demonstrated that highly committed efforts can increase screening rates, reduce incidence rates, eliminate racial disparities in outcomes, and reduce cost.148 Vaccination against viral causes of GI cancer such as hepatitis B and C and HPV as well as the eradication of H pylori, if widely applied in high-risk regions, have the potential to eliminate or substantially decrease the incidence of these diseases where they are endemic but will require concerted public health efforts in many countries to achieve these goals. As is the case with most cancers, it is the interplay of basic science research, robust clinical trials, and dissemination of known effective treatments to all communities that will enable continued progress against GI cancers during ASCO’s next 50 years. AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Although all authors completed the disclosure declaration, the following author(s) and/or an author’s immediate family member(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO’s conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors. Employment or Leadership Position: None Consultant or Advisory Role: Alan P. Venook, Bristol-Myers Squibb (U), Genentech/Roche (U) Stock Ownership: None Honoraria: None Research Funding: Alan P. Venook, GlaxoSmithKline, Genentech, Genomic Health Expert Testimony: None Patents, Royalties, and Licenses: None Other Remuneration: None AUTHOR CONTRIBUTIONS

Administrative support: Robert J. Mayer Manuscript writing: All authors Final approval of manuscript: All authors REFERENCES 1. [No authors listed]: Statistics on cancer. CA Cancer J Clin 17:34-43, 1967 2. Siegel R, Ma J, Zou Z: Cancer statistics, 2014. CA Cancer J Clin 64:9-29, 2014 3. Jemal A, Bray F, Center MM, et al: Global cancer statistics. CA Cancer J Clin 61:69-90, 2011 4. Moertel CG, Childs DS Jr, Reitemeier RJ, et al: Combined 5-fluorouracil and supervoltage radiation therapy of locally unresectable gastrointestinal cancer. Lancet 2:865-867, 1969 5. Blot WJ, Devesa SS, Kneller RW, et al: Rising incidence of adenocarcinoma of the esophagus and gastric cardia. JAMA 265:1287-1289, 1991 6. Wynder EL, Mabuchi K: Cancer of the gastrointestinal tract: Etiological and environmental factors. JAMA 226:1546-1548, 1973 7. Cook MB, Chow WH, Devesa SS: Oesophageal cancer incidence in the United States by race, sex, and histologic type, 1977-2005. Br J Cancer 101:855-859, 2009 www.jco.org

8. Lagergren J, Bergstro¨m R, Lindgren A, et al: Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 340:825-831, 1999 9. Spechler SJ: Barrett esophagus and risk for esophageal cancer: A clinical review. JAMA 310:627-636, 2013 10. Turati F, Tramacere I, La Vecchia C, et al: A meta-analysis of body mass index and esophageal and gastric cardia adenocarcinoma. Ann Oncol 24:609-617, 2013 11. Reid BJ, Li X, Galipeau PC, et al: Barrett’s esophagus and oesophageal adenocarcinoma: Time for a new synthesis. Nat Rev Cancer 10:87-101, 2010 12. Moertel CG: Alimentary tract cancer: The esophagus, in Holland JR, Frei E III (eds): Cancer Medicine. Lea & Febiger, Philadelphia, PA, 1973, p1523 13. Biere SS, van Berge Henegouwen MI, Maas KW, et al: Minimally invasive versus open oesophagectomy for patients with oesophageal cancer: A multicenter, open-label, randomized controlled trial. Lancet 379:1887-1892, 2012 14. Sjoquist KM, Burmeister BH, Smithers BM, et al: Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: An updated metaanalysis. Lancet Oncol 12:681-692, 2011 15. van Hagen P, Hulshof MC, van Lanschot JJ, et al: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366:2074-2084, 2012 16. Bedenne L, Michel P, Bouche´ O, et al: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25:1160-1168, 2007 17. Stahl M, Stuschke M, Lehmann N, et al: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23:2310-2317, 2005 18. Lordick F, Ott K, Krause BJ, et al: PET to assess early metabolic response and to guide treatment of adenocarcinoma of the oesophagogastric junction: The MUNICON phase II trial. Lancet Oncol 8:797-805, 2007 19. Wiggins CL, Becker TM, Key CR, et al: Stomach cancer among New Mexico’s American Indians, Hispanic whites, and non-Hispanic whites. Cancer Res 49:1595-1599, 1989 20. Hanley AJ, Choi BC, Holowaty EJ: Cancer mortality among Chinese migrants: A review. Int J Epidemiol 24:255-265, 1995 21. Chyou PH, Nomurg AM, Hanker JH, et al: A case-cohort study of diet and stomach cancer. Cancer Res 50:7501-7504, 1990 22. Uemura N, Okamoto S, Yamamoto S, et al: Helicobacter pylori infection and the development of gastric cancer. N Engl J Med 345:784-789, 2001 23. Lauren P: The two histological main types of gastric carcinoma: Diffuse and so-called intestinal-type carcinomas—An attempt at histo-clinical classification. Acta Pathol Microbiol Scand 64:31-49, 1965 24. Kaurah P, MacMillan A, Boyd N, et al: Founder and recurrent CDH1 mutations in families with hereditary diffuse gastric cancer. JAMA 297:23602372, 2007 25. Bozzetti F, Marubini E, Bonfanti G, et al: Subtotal versus total gastrectomy for gastric cancer: Five-year survival rates in a multicenter randomized Italian trial—Italian Gastrointestinal Tumor Study Group. Ann Surg 230:170-178, 1999 26. Songun I, Putter H, Kranenberg EM, et al: Surgical treatment of gastric cancer: 15 year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol 11:439-449, 2010 27. Power DG, Kelsen DP, Shah MA: Advanced gastric cancer: Slow but steady progress. Cancer Treat Rev 36:384-392, 2010 28. Bang YJ, Van Cutsem E, Feyereislova A, et al: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomized controlled trial. Lancet 376:687-697, 2010 29. Ohtsu A, Shah MA, Van Cutsem E, et al: Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: A randomized double-blind, placebo-controlled phase III study. J Clin Oncol 29:3968-3976, 2011 30. Fuchs CS, Tomasek J, Yong CJ, et al: Ramucirurab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): An international randomised multicentre, placebo-controlled, phase 3 trial. Lancet 383:31-39, 2014 31. Wilke H, Van Cutsem E, Oh SC, et al: RAINBOW: A global, phase III, randomized, double-blind study of ramucirurab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastroesophageal junction (GEJ) and gastric adenocarcinoma following disease progression on first-line platinumand fluoropyrimidine-containing combination therapy rainbow IMCL CP12-0922 (I4T-IE-JVBE). J Clin Oncol 32, 2014 (suppl 3; abstr LBA7) 32. Bang YJ, Kim YW, Yang HK, et al: Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): A phase 3 open-label, randomised controlled trial. Lancet 379:315-321, 2012

© 2014 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

1527

Mayer, Venook, and Schilsky

33. Sasako M, Sakuramoto S, Katai H, et al: Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol 29:4387-4393, 2011 34. Cunningham D, Allum WH, Stenning SP, et al: Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 355:11-20, 2006 35. Smalley SR, Benedetti JK, Haller DG, et al: Updated analysis of SWOGdirected Intergroup study 0116: A phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol 30:23272333, 2012 36. Fuchs CS, Tepper JE, Niedzwiecki D, et al: Postoperative adjuvant chemoradiation for gastric or gastroesophageal junction (GEJ) adenocarcinoma using epirubicin, cisplatin, and infusional (CI) 5-FU (ECF) before and after CI 5-FU and radiotherapy (CRT) compared with bolus 5-FU/LV before and after CRT: Intergroup trial CALGB 80101. J Clin Oncol 29:45s, 2011 (suppl; abstr 4003) 37. Edis AJ, Kiernan PD, Taylor WF: Attempted curative resections of ductal carcinoma of the pancreas: Review of Mayo Clinic experience, 1951-1975. Mayo Clin Proc 55:531-536, 1980 38. McCune WS, Shorb PE, Moscovitz H: Endoscopic cannulation of the ampulla of Vater: A preliminary report. Ann Surg 167:752-756, 1968 39. Faigel DO, Ginsberg GG, Bentz JS, et al: Endoscopic ultrasound-guided real-time fine-needle aspiration biopsy of the pancreas in cancer patients with pancreatic lesions. J Clin Oncol 15:1439-1443, 1997 40. Yeo CJ, Cameron JL, Sohn TA, et al: Six hundred fifty consecutive pancreaticoduodenectomies in the 1990s: Pathology, complications, and outcomes. Ann Surg 226:248-260, 1997 41. Feig C, Gopinathan A, Neesse A, et al: The pancreas cancer microenvironment. Clin Cancer Res 18:4266-4276, 2012 42. Jones S, Zhang X, Parsons DW, et al: Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321:18011806, 2008 43. Pleskow DK, Berger HJ, Gyves J, et al: Evaluation of a serologic marker, CA19-9, in the diagnosis of pancreatic cancer. Ann Intern Med 110:704-709, 1989 44. Hruban RH, Adsay NV, Albores-Saavedra J, et al: Pathology of genetically engineered mouse models of pancreatic exocrine cancer: Consensus report and recommendations. Cancer Res 66:95-106, 2006 45. Li D, Morris JS, Liu J, et al: Body mass index and risk, age of onset, and survival in patients with pancreatic cancer. JAMA 301:2553-2562, 2009 46. Gullo L, Pezzilli R, Morselli-Labate AM, et al: Diabetes and the risk of pancreatic cancer. N Engl J Med 331:81-84, 1994 47. Fuchs CS, Colditz GA, Stamfer MJ, et al: A prospective study of cigarette smoking and the risk of pancreatic cancer. Arch Intern Med 156:2255-2260, 1996 48. Shi C, Hruban RH, Klein AP: Familial pancreatic cancer. Arch Pathol Lab Med 133:365-374, 2009 49. Kastrinos F, Mukherjee B, Tayob N, et al: Risk of pancreatic cancer in families with Lynch syndrome. JAMA 302:1790-1795, 2009 50. Couch FJ, Johnson MR, Rabe KG, et al: The prevalence of BRCA2 mutations in familial pancreatic cancer. Cancer Epidemiol Biomarkers Prev 16:342-346, 2007 51. Jones J, Hruban RH, Kamiyama M, et al: Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 324:217, 2009 52. Kalser MH, Ellenberg SS: Pancreatic cancer: Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 120:899-903, 1985 53. Burris HA III, Moore MJ, Andersen J, et al: Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol 15:2403-2413, 1997 54. Oettle H, Neuhaus P, Hochhaus A, et al: Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: The CONKO-001 randomized trial. JAMA 310:1473-1481, 2013 55. Neoptolemos JP, Stocken DD, Friess H, et al: A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 350:1200-1210, 2004 56. Klinkenbijl JH, Jeekel J, Sahmoud T: Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: Phase III trial of the EORTC Gastrointestinal Tract Cancer Cooperative Group. Ann Surg 230:776-784, 1999 57. Regine WF, Winter KA, Abrams RA, et al: Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: A randomized controlled trial. JAMA 299:1019-1026, 2008 58. Hidalgo M: Pancreatic cancer. N Engl J Med 362:1605-1617, 2010 59. Conroy T, Desseigne F, Ychou M, et al: FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer: N Engl J Med 364:1817-1825, 2011 1528

© 2014 by American Society of Clinical Oncology

60. Gourgou-Bourgade S, Bascoul-Mollevi C, Desseigne F, et al: Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: Results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol 31:23-29, 2013 61. Von Hoff DD, Ervin T, Arena FP, et al: Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369:1691-1703, 2013 62. Beasley RP, Hwang LY, Lin CC, et al: Hepatocellular carcinoma and hepatitis B virus: A prospective study of 22707 men in Taiwan. Lancet 2:11291133, 1981 63. Chang DT, Schellenberg D, Shen J, et al: Stereotactic radiotherapy for unresectable adenocarcinoma of the pancreas. Cancer 115:665-672, 2009 64. El-Serag HB, Mason AC: Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med 340:745-750, 1999 65. Mazzaferro V, Regalia E, Doci R, et al: Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med 334:693-699, 1996 66. Yao FY, Ferrell L, Bass NM, et al: Liver transplantation for hepatocellular carcinoma: Expansion of the tumor size limits does not adversely impact survival. Hepatology 33:1394-1403, 2001 67. US Department of Health and Human Services: Organ and Transplantation Network: Transplant annual data reports 2012. http://optn.transplant.hrsa .gov/data 68. Llovet JM, Real MI, Montan˜a X, et al: Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: A randomised controlled trial. Lancet 359:1734-1739, 2002 69. Cheng A, Kang Y, Chen Z, et al: Randomized phase III trial of sorafenib versus placebo in Asian patients with advanced hepatocellular carcinoma. J Clin Oncol 26:215s, 2008 (suppl; abstr A4509) 70. Llovet JM, Ricci S, Mazzaferro V, et al: Sorabenib in advanced hepatocullular carcinoma. N Engl J Med 359:378-390, 2008 71. Llovet JM, Di Bisceglie AM, Bruiz J: Design and endpoints of clinical trials in hepatocellular carcinoma. J Natl Cancer Inst 100:698-711, 2008 72. Valle J, Wasan H, Palmer DH, et al: Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med 362:1273-1281, 2010 73. Brazeau P, Vale W, Burgus R, et al: Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science 179:77-79, 1973 74. Kvols LK: Metastatic carcinoid tumors and the carcinoid syndrome: A selective review of chemotherapy and hormonal therapy. Am J Med 81:49-55, 1986 75. Rubin J, Ajani J, Schirmer W, et al: Octreotide acetate long-acting formulation versus open-label subcutaneous octreotide acetate in malignant carcinoid syndrome. J Clin Oncol 17:600-606, 1999 76. Raymond E, Dahan L, Raoul JL: Sunitinib maleate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 364:501-513, 2011 77. Yao JC, Shah MH, Ito T, et al: Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364:514-523, 2011 78. Chan DS, Lau R, Aune D, et al: Red and processed meat and colorectal cancer incidence: Meta-analysis of prospective studies. PLoS One 6:e20456, 2011 79. Bardou M, Barkun AN, Martel M: Obesity and colorectal cancer. Gut 62:933-947, 2013 80. Eheman C, Henley SJ, Ballard-Barbash R, et al: Annual report to the nation on the status of cancer, 1975-2008, featuring cancers associated with excess weight and lack of sufficient physical activity. Cancer 118:2338-2366, 2012 81. Rothwell PM, Wilson M, Elwin CE, et al: Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet 376:1741-1750, 2010 82. Chan AT, Arber N, Burn J, et al: Aspirin in the chemoprevention of colorectal neoplasia: An overview. Cancer Prev Res (Phila) 5:164-178, 2012 83. Chan AT, Ogino S, Fuchs CS: Aspirin use and survival after diagnosis of colorectal cancer. JAMA 302:649-658, 2009 84. Meyerhardt JA, Giovannucci EL, Holmes MD, et al: Physical activity and survival after colorectal cancer diagnosis. J Clin Oncol 24:3527-3534, 2006 85. Patel SG, Ahnen DJ: Familial colon cancer syndromes: An update of a rapidly evolving field. Curr Gastroenterol Rep 14:428-438, 2012 86. Peipins LA, Sandler RS: Epidemiology of colorectal adenomas. Epidemiol Rev 16:273-297, 1994 87. Shaukat A, Mongin SJ, Geisser MS, et al: Long-term mortality after screening for colorectal cancer. N Engl J Med 369:1106-1114, 2013 88. Atkin WS, Edwards R, Kralj-Hans I, et al: Once-only flexible sigmoidoscopy screening in prevention or colorectal cancer: A multicentre randomised controlled trial. Lancet 375:1624-1633, 2010 JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

ASCO 50th Anniversary

89. Schoen RE, Pinsky PF, Weissfeld JL, et al: Colorectal cancer incidence and mortality with screening flexible sigmoidoscopy. N Engl J Med 366:23452357, 2012 90. Johnson CD, Chen M-H, Toledano AY, et al: Accuracy of CT colonography for detection of large adenomas and cancers. N Engl J Med 359:1207-1217, 2008 91. Imperiale TF, Ransohoff DF, Itzkowitz SH, et al: Fecal DNA versus fecal occult blood for colorectal cancer screening in an average-risk population. N Engl J Med 351:2704-2714, 2004 92. Vogelstein B, Fearon ER, Hamilton SR, et al: Genetic alterations during colorectal-tumor development. N Engl J Med 319:525-532, 1988 93. Markowitz SD, Bertagnolli MM: Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med 361:2449-2460, 2009 94. The Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature 487:330-337, 2012 95. Edge SB, Byrd DR, Compton CC, et al (eds): Cancer Staging Handbook from the AJCC Cancer Staging Manual (ed 7). Springer, New York, NY, 2010 96. Chang GJ, Rodriguez-Bigas MA, Skibber JM, et al: Lymph node evaluation and survival after curative resection of colon cancer: Systemic review. J Natl Cancer Inst 99:433-441, 2007 97. Johnson PM, Porter GA, Ricciardi R, et al: Increasing negative lymph node count is independently associated with improved long-term survival in stage IIIB and IIIC colon cancer. J Clin Oncol 24:3570-3575, 2006 98. Wolmark N, Fisher B, Wieand HS, et al: The prognostic significance of preoperative carcinoembryonic antigen levels in colorectal cancer: Results from NSABP (National Surgical Adjuvant Breast and Bowel Project) clinical trials. Ann Surg 199:375-382, 1984 99. Popat S, Hubner R, Houlston RS: Systemic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol 23:609-618, 2005 100. Sargent DJ, Marsoni S, Monges G, et al: Defective mismatch repair as a predictive marker for lack of efficiency of fluorouracil-based therapy in colon cancer. J Clin Oncol 28:3219-3226, 2010 101. Roth AD, Tejpar S, Delorenzi M, et al: Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: Results of the translational study of the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol 28:466-474, 2009 102. Ogino S, Shima K, Meyerhardt JA, et al: Predictive and prognostic roles of BRAF mutation in stage III colon cancer: Results from Intergroup trial CALGB 89803. Clin Cancer Res 18:890-900, 2012 103. Salazar R, Roepman P, Capella G, et al: Gene expression signature to improve prognosis prediction of stage II and III colorectal cancer. J Clin Oncol 29:17-24, 2011 104. Venook AP, Niedzwiecki D, Lopatin M, et al: Biological determinants of tumor recurrence in stage II colon cancer: Validation study of the 12-gene recurrence score in Cancer and Leukemia Group B (CALGB) 9581. J Clin Oncol 31:1775-1781, 2013 105. Yothers G, O’Connell MJ, Lee M, et al: Validation of the 12-gene colon cancer recurrence score in NSABP C-07 as a predictor of recurrence in patients with stage II and III colon cancer treated with fluorouracil and leucovorin (FU/LV) and FU/LV plus oxaliplatin. J Clin Oncol 31:4512-4519, 2014 106. Di B, Li Y, Wei K, et al: Laparoscopic versus open surgery for colon cancer: A meta-analysis of 5-year follow-up outcomes. Surg Oncol 22:e39-e43, 2013 107. Andre´ T, Boni C, Navarro M, et al: Improved overall survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III colon cancer in the MOSAIC trial. J Clin Oncol 27:3109-3116, 2009 108. Saltz LB, Niedzwiecki D, Hollis D, et al: Irinotecan, fluorouracil plus leucovorin is not superior to fluorouracil plus leucovorin alone as adjuvant treatment for stage III colon cancer: Results of CALGB 89803. J Clin Oncol 25:3456-3461, 2007 109. Van Cutsem E, Labianca R, Bodoky G, et al: Randomized phase III trial comparing biweekly infusional fluorouracil/leucovorin alone or with irinotecan in the adjuvant treatment of stage III colon cancer: PETACC-3. J Clin Oncol 27:3117-3125, 2009 110. Allegra CJ, Yothers G, O’Connell MJ, et al: Phase III trial assessing bevacizumab in stages II and III carcinoma of the colon: Results of NSABP protocol C-08. J Clin Oncol 29:11-16, 2011 111. Alberts SR, Sargent DJ, Nair S, et al: Effect of oxaliplatin, fluorouracil and leucovorin with or without cetuximab on survival among patients with resected stage III colon cancer. A randomized trial. JAMA 307:1383-1393, 2012 112. Kapiteijn E, Marijnen CA, Nagtegaal ID, et al: Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med 345:638-646, 2001 113. Ro¨del C, Liersch T, Becker H, et al: Preoperative chemoradiotherapy and postoperative chemotherapy with fluorouracil and oxaliplatin versus fluorouracil alone in locally advanced rectal cancer: Initial results of the German CAO/ARO/ A10-04 randomised phase 3 trial. Lancet Oncol 13:679-687, 2012 www.jco.org

114. Sauer R, Liersch T, Merkel S, et al: Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: Results of the German CAO/ARO/AR94 randomized phase III trial after a median follow-up of 11 years. J Clin Oncol 30:1926-1933, 2012 115. Schrag D, Cramer LD, Bach PB, et al: Influence of hospital procedure volume on outcomes following surgery for colon cancer. JAMA 284:3028-3035, 2000 116. Locker GY, Hamilton S, Harris J, et al: ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol 24:5313-5327, 2006 117. Wong SL, Mangu PB, Choti MA, et al: American Society of Clinical Oncology 2009 clinical evidence review on radiofrequency ablation of hepatic metastases from colorectal cancer. J Clin Oncol 28:493-508, 2010 118. Advanced Colorectal Cancer Meta-Analysis Project: Modulation of fluorouracil by leucovorin in patients with advanced colorectal cancer: Evidence in terms of response rate. J Clin Oncol 10:896-903, 1992 119. Douillard JY, Cunningham D, Roth AD, et al: Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: A multicenter randomised trial. Lancet 355:1041-1047, 2000 120. DeGramont A, Figer A, Seymour M, et al: Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 18:2983-2947, 2000 121. Tournigand C, Andre´ T, Achille E, et al: FOLFIRI followed by FOLFOX 6 or the reverse sequence in advanced colorectal cancer: A randomized GERCOR study. J Clin Oncol 22:229-237, 2004 122. Van Cutsem E, Twelves C, Cassidy J, et al: Oral capecitabine compared with intravenous fluorouracil plus leucovorin in patients with metastatic colorectal cancer: Results of a large phase III study. J Clin Oncol 19:4097-4106, 2001 123. Twelves C, Wong A, Nowacki MP, et al: Capecitabine as adjuvant treatment for stage III colon cancer. N Engl J Med 352:2696-2704, 2005 124. Hurwitz H, Fehrenbacher L, Novotny W, et al: Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350:2335-2342, 2004 125. Hurwitz HI, Tebbutt NC, Kabbinavar F, et al: Efficacy and safety of bevacizumab in metastatic colorectal cancer: Pooled analysis from seven randomized controlled trials. Oncologist 18:1004-1012, 2013 126. Cunningham D, Humblet Y, Siena S, et al: Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 351:337-345, 2004 127. Van Cutsem E, Ko¨hne CH, La´ng I, et al: Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: Updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 29:2011-2019, 2011 128. Dahabreh IJ, Terasawa T, Castaldi PJ, et al: Systematic review: Anti– epidermal growth factor receptor treatment effect modification by KRAS mutations in advanced colorectal cancer. Ann Intern Med 154:37-49, 2011 129. Douillard J-Y, Oliner KS, Siena S, et al: Panitumumab–FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 369:1023-1034, 2013 130. Allegra CJ, Jessup JM, Somerfield MR, et al: American Society of Clinical Oncology provisional clinical opinion: Testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti– epidermal growth factor receptor monoclonal antibody therapy. J Clin Oncol 27:2091-2096, 2009 131. Hecht JR, Mitchell E, Chidiac T, et al: A randomized phase IIIB trial of chemotherapy, bevacizumab, and panitumumab compared with chemotherapy and bevacizumab alone in metastatic colorectal cancer. J Clin Oncol 27:627-680, 2009 132. Tol J, Koopman M, Cats A, et al: Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. N Engl J Med 360:563-572, 2009 133. Heinemann V, von Weikersthal LF, Decker T, et al: Randomized comparison of FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment of KRAS wild-type metastatic colorectal cancer: German AIO study KRK-0306 (FIRE-3). J Clin Oncol 31:206s, 2013 (suppl 15; abstr LBA3506) 134. Van Cutsem E, Tabernero J, Lakomy R, et al: Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol 30:3499-3506, 2012 135. Grothey A, Van Cutsem E, Sobrero A, et al: Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): An international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 381:303-312, 2013 136. Holly EA, Whittemore AS, Aston DA, et al: Anal cancer incidence: Genital warts, anal fissure or fistula, hemorrhoids, and smoking. J Natl Cancer Inst 81:1726-1731, 1989 137. Beahrs OH, Wilson SM: Carcinoma of the anus. Ann Surg 184:422-428, 1976

© 2014 by American Society of Clinical Oncology

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

1529

Mayer, Venook, and Schilsky

138. Frisch M, Glimelius B, van den Brule AJ, et al: Sexually transmitted infection as a cause of anal cancer. N Engl J Med 337:1350-1358, 1997 139. Daling JR, Weiss NS, Klopfenstein LL, et al: Correlates of homosexual behavior and the incidence of anal cancer. JAMA 247:1988-1990, 1982 140. Melbye M, Cote´ TR, Kessler L, et al: High incidence of anal cancer among AIDS patients: The AIDS/Cancer Working Group. Lancet 343:636-639, 1994 141. Palefsky JM, Giuliano AR, Goldstone S, et al: HPV vaccine against anal HPV infection and anal intraepithelial neoplasia. N Engl J Med 365:1576-1585, 2011 142. Nigro ND, Vaitkevicius VK, Considine B Jr: Combined therapy for cancer of the anal canal: A preliminary report. Dis Colon Rectum 17:354-356, 1974 143. Leichman L, Nigro N, Vaitkevicius VK, et al: Cancer of the anal canal: Model for preoperative adjuvant combined modality therapy. Am J Med 78:211215, 1985 144. Bartelink H, Roelofsen F, Eschwege F, et al: Concomitant radiotherapy and chemotherapy is superior to radiotherapy alone in the treatment of locally advanced anal cancer: Results of a phase III randomized trial of the European Organisation for Research and Treatment of Cancer Radiotherapy and Gastrointestinal Cooperative Groups. J Clin Oncol 15:2040-2049, 1997

145. UKCCCR Anal Cancer Trial Working Party: Epidermoid anal cancer: Results from the UKCCCR randomised trial of radiotherapy alone versus radiotherapy, 5-fluorouracil, and mitomycin. Lancet 348:1049-1054, 1996 146. Flam M, John M, Pajak TF, et al: Role of mitomycin in combination with fluorouracil and radiotherapy, and of salvage chemoradiation in the definitive nonsurgical treatment of epidermoid carcinoma of the anal canal: Results of a phase III randomized intergroup study. J Clin Oncol 14:2527-2539, 1996 147. Gunderson LL, Winter KA, Ajani JA, et al: Long-term update of US GI Intergroup RTOG 98-11 phase III trial for anal carcinoma: Survival, relapse, and colostomy failure with concurrent chemoradiation involving fluorouracil/mitomycin versus fluorouracil/cisplatin. J Clin Oncol 30:4344-4351, 2012 148. Grubbs SS, Polite BN, Carney J Jr, et al: Eliminating racial disparities in colorectal cancer in the real world: It took a village. J Clin Oncol 31:1928-1930, 2013

DOI: 10.1200/JCO.2014.55.4121; published online ahead of print at www.jco.org on April 21, 2014

■ ■ ■

ASCO Celebrates 50 Years of Advancing Progress Against Cancer This historic year, as ASCO proudly commemorates its 50th anniversary and decades of evolutionary growth, the Society also celebrates the significant progress that has been made against cancer throughout history. ASCO’s anniversary website, CancerProgress.Net, chronicles these achievements and more. We invite you to visit the upgraded Cancer Progress Timeline to explore advances in 18 different cancers and several types of care, peruse stories about ASCO’s evolution and progress in the field, check out the site’s new social media features, and vote on the most significant milestones in the field. You can also follow ASCO on ASCO Connection, Twitter, and Facebook to join in on the conversation about progress.

1530

© 2014 by American Society of Clinical Oncology

JOURNAL OF CLINICAL ONCOLOGY

Downloaded from jco.ascopubs.org on October 4, 2014. For personal use only. No other uses without permission. Copyright © 2014 American Society of Clinical Oncology. All rights reserved.

Progress against GI cancer during the American Society of Clinical Oncology's first 50 years.

Progress against GI cancer during the American Society of Clinical Oncology's first 50 years. - PDF Download Free
218KB Sizes 0 Downloads 3 Views