16.01.2013 Views

Practice Guidelines in Oncology - Gastric Cancer

Practice Guidelines in Oncology - Gastric Cancer

Practice Guidelines in Oncology - Gastric Cancer

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

NCCN Cl<strong>in</strong>ical <strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong> <strong>in</strong> <strong>Oncology</strong><br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

V.1.2007<br />

Cont<strong>in</strong>ue<br />

www.nccn.org


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

* Jaffer Ajani, MD/Chair †¤<br />

The University of Texas M. D. Anderson<br />

<strong>Cancer</strong> Center<br />

* Tanios Bekaii-Saab, MD †<br />

Arthur G. James <strong>Cancer</strong> Hospital &<br />

Richard J. Solove Research Institute at<br />

The Ohio State University<br />

* Thomas A. D’Amico, MD <br />

Duke Comprehensive <strong>Cancer</strong> Center<br />

Charles Fuchs, MD †<br />

Dana-Farber/Brigham and Women’s<br />

<strong>Cancer</strong> Center | Massachusetts General<br />

Hospital <strong>Cancer</strong> Center<br />

Michael K. Gibson, MD †Þ<br />

The Sidney Kimmel Comprehensive<br />

<strong>Cancer</strong> Center at Johns Hopk<strong>in</strong>s<br />

Melvyn Goldberg, MD <br />

Fox Chase <strong>Cancer</strong> Center<br />

James A. Hayman, MD, MBA §<br />

University of Michigan Comprehensive<br />

<strong>Cancer</strong> Center<br />

† Medical oncology<br />

¤ Gastroenterology<br />

Surgery/Surgical oncology<br />

Þ Internal medic<strong>in</strong>e<br />

§ Radiotherapy/Radiation oncology<br />

‡ Hematology/Hematology oncology<br />

*Writ<strong>in</strong>g committee member<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

NCCN <strong>Gastric</strong> <strong>Cancer</strong> Panel Members<br />

David H. Ilson, MD, PhD †Þ<br />

Memorial Sloan-Ketter<strong>in</strong>g <strong>Cancer</strong> Center<br />

Mil<strong>in</strong>d Javle, MD †<br />

Roswell Park <strong>Cancer</strong> Institute<br />

Scott T. Kelley, MD <br />

H. Lee Moffitt <strong>Cancer</strong> Center and Research<br />

Institute at the University of South Florida<br />

Robert C. Kurtz, MD ¤Þ<br />

Memorial Sloan-Ketter<strong>in</strong>g <strong>Cancer</strong> Center<br />

Gershon Yehuda Locker, MD †<br />

Robert H. Lurie Comprehensive <strong>Cancer</strong><br />

Center at Northwestern University<br />

Neal J. Meropol, MD †<br />

Fox Chase <strong>Cancer</strong> Center<br />

Bruce D. M<strong>in</strong>sky, MD §<br />

Memorial Sloan-Ketter<strong>in</strong>g <strong>Cancer</strong> Center<br />

Mark B. Orr<strong>in</strong>ger, MD <br />

University of Michigan Comprehensive<br />

<strong>Cancer</strong> Center<br />

Cont<strong>in</strong>ue<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Raymond U. Osarogiagbon, MD †Þ‡<br />

St. Jude Children’s Research<br />

Hospital/University of Tennessee <strong>Cancer</strong><br />

Institute<br />

James A. Posey, MD †<br />

University of Alabama at Birm<strong>in</strong>gham<br />

Comprehensive <strong>Cancer</strong> Center<br />

Jack Roth, MD <br />

The University of Texas M.D. Anderson<br />

<strong>Cancer</strong> Center<br />

* Aaron R. Sasson, MD <br />

UNMC Eppley <strong>Cancer</strong> Center at The<br />

Nebraska Medical Center<br />

Stephen G. Swisher, MD <br />

The University of Texas M. D. Anderson<br />

<strong>Cancer</strong> Center<br />

Douglas E. Wood, MD <br />

Fred Hutch<strong>in</strong>son <strong>Cancer</strong> Research<br />

Center/Seattle <strong>Cancer</strong> Care Alliance<br />

Gary Yang, MD §<br />

Roswell Park <strong>Cancer</strong> Institute<br />

Yun Yen, MD, PhD ‡<br />

City of Hope <strong>Cancer</strong> Center


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Table of Contents<br />

NCCN <strong>Gastric</strong> <strong>Cancer</strong> Panel Members<br />

Workup and Evaluation (GAST-1)<br />

Postlaparoscopy Stag<strong>in</strong>g and Treatment (GAST-2)<br />

Adjunctive Treatment (GAST-3)<br />

Follow-up and Salvage Therapy (GAST-4)<br />

Pr<strong>in</strong>ciples of Surgery (GAST-A)<br />

Pr<strong>in</strong>ciples of Systemic Therapy (GAST-B)<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

Pr<strong>in</strong>t the <strong>Gastric</strong> <strong>Cancer</strong> Guidel<strong>in</strong>e<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

For help us<strong>in</strong>g these<br />

documents, please click here<br />

Stag<strong>in</strong>g<br />

Manuscript<br />

References<br />

This manuscript is be<strong>in</strong>g<br />

updated to correspond<br />

with the newly updated<br />

algorithm.<br />

Cl<strong>in</strong>ical Trials: The NCCN<br />

believes that the best management<br />

for any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical<br />

trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is<br />

especially encouraged.<br />

To f<strong>in</strong>d cl<strong>in</strong>ical trials onl<strong>in</strong>e at NCCN<br />

member <strong>in</strong>stitutions, click here:<br />

nccn.org/cl<strong>in</strong>ical_trials/physician.html<br />

NCCN Categories of Consensus:<br />

All recommendations are Category<br />

2A unless otherwise specified.<br />

See NCCN Categories of Consensus<br />

Summary of <strong>Guidel<strong>in</strong>es</strong> Updates<br />

These guidel<strong>in</strong>es are a statement of consensus of the authors regard<strong>in</strong>g their views of currently accepted approaches to treatment. Any cl<strong>in</strong>ician<br />

seek<strong>in</strong>g to apply or consult these guidel<strong>in</strong>es is expected to use <strong>in</strong>dependent medical judgment <strong>in</strong> the context of <strong>in</strong>dividual cl<strong>in</strong>ical circumstances to<br />

determ<strong>in</strong>e any patient's care or treatment. The National Comprehensive <strong>Cancer</strong> Network makes no representations nor warranties of any k<strong>in</strong>d<br />

whatsoever regard<strong>in</strong>g their content, use, or application and disclaims any responsibility for their application or use <strong>in</strong> any way. These guidel<strong>in</strong>es are<br />

copyrighted by National Comprehensive <strong>Cancer</strong> Network. All rights reserved. These guidel<strong>in</strong>es and the illustrations here<strong>in</strong> may not be reproduced <strong>in</strong><br />

any form without the express written permission of NCCN. ©2007.


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Summary of the <strong>Guidel<strong>in</strong>es</strong> Updates<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Summary of major changes <strong>in</strong> the 1.2007 version of the <strong>Gastric</strong> <strong>Cancer</strong> guidel<strong>in</strong>es from the 2006 version <strong>in</strong>clude:<br />

( GAST-1):<br />

� Additional Evaluation: Laparoscopic recommendations were condensed and changed to “Consider laparoscopy (category 2B)”<br />

� Added new footnote “a” regard<strong>in</strong>g the appropriateness of PET/CT scans for T1 or M1 patients<br />

( GAST-2):<br />

� Medically fit, potentially resectable: M0 pathway revised to <strong>in</strong>clude T1 and T2 tumors<br />

� Added new footnote “d” about surgery as primary treatment for treatment for T1 cancer<br />

( GAST-A)<br />

� A new page entitled, “Pr<strong>in</strong>ciples of Surgery” was added to outl<strong>in</strong>e recommended guidel<strong>in</strong>es for gastric surgery<br />

( GAST-B):<br />

� All category of consensus recommendations for systemic therapies were revised to reflect current data<br />

� Preoperative Chemotherapy: Added “ECF”<br />

� Postoperative Chemotherapy: Added “ECF”<br />

� “5-FU” was changed to “fluoropyrimid<strong>in</strong>e” throughout page<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

UPDATES


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

WORKUP<br />

� Multidiscipl<strong>in</strong>ary<br />

evaluation<br />

� H&P<br />

� CBC, platelets, SMA-12<br />

� Abdom<strong>in</strong>al CT<br />

� CT/ultrasound pelvis<br />

(females)<br />

� Chest x-ray<br />

� Esophagogastroduodenoscopy<br />

� PET/CT scana a<br />

b<br />

c<br />

CLINICAL<br />

PRESENTATION<br />

Locoregional<br />

(M0)<br />

Stage IV<br />

(M1)<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Medically fit, b<br />

potentially<br />

resectable<br />

Medically fit, b<br />

unresectable<br />

Medically unfit<br />

May not be appropriate for T1 or M1 patients.<br />

Medically able to tolerate major abdom<strong>in</strong>al surgery.<br />

Laparoscopy is performed to evaluate for peritoneal spread when consider<strong>in</strong>g chemotherapy/RT or surgery.<br />

Laparoscopy is not <strong>in</strong>dicated if a palliative resection is planned.<br />

ADDITIONAL<br />

EVALUATION<br />

Cosider<br />

Laparoscopyc<br />

(category 2B)<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Postlaparoscopy<br />

Stag<strong>in</strong>g (see GAST-2)<br />

Salvage Therapy<br />

(see GAST-4)<br />

GAST-1


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Medically fit, b<br />

potentially<br />

resectable<br />

Medically fit, b<br />

unresectable<br />

Medically<br />

unfit<br />

b<br />

d<br />

POSTLAPAROSCOPY<br />

STAGING<br />

M0<br />

M1<br />

M0<br />

M1<br />

M0<br />

M1<br />

T1 or less<br />

(by cl<strong>in</strong>ical stag<strong>in</strong>g)<br />

T2 or higher<br />

(by cl<strong>in</strong>ical<br />

stag<strong>in</strong>g or N+)<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

PRIMARY<br />

TREATMENT<br />

Surgery d,e<br />

Surgery<br />

RT, 45-50.4 Gy + concurrent<br />

5-FU-based radiosensitization (category 1)<br />

or Chemotherapy f<br />

Salvage Therapy<br />

(see GAST-4)<br />

RT, 45–50.4 Gy + concurrent<br />

5-FU-based radiosensitization<br />

(category 1)<br />

or<br />

Salvage Therapy<br />

(see GAST-4)<br />

Salvage Therapy<br />

(see GAST-4)<br />

Medically able to tolerate major abdom<strong>in</strong>al surgery.<br />

Surgery as primary therapy is appropriate for T1 cancer or actively bleed<strong>in</strong>g cancer, or when postoperative adjuvant therapy is preferred.<br />

eSee<br />

Pr<strong>in</strong>ciples of Surgery (GAST-A) .<br />

fSee<br />

Pr<strong>in</strong>ciples of Systemic Therapy (GAST-B) .<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

or<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

e<br />

Preoperative<br />

chemotherapyf<br />

Salvage Therapy<br />

(see GAST-4)<br />

Surgery e<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Surgical Outcomes<br />

(see GAST-3)<br />

Adjunctive Treatment<br />

Postchemotherapy ± RT<br />

(see GAST-3)<br />

Adjunctive Treatment<br />

Postchemotherapy ± RT<br />

(see GAST-3)<br />

GAST-2


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Surgical<br />

outcomes<br />

SURGICAL RESECTION<br />

R0 resection<br />

R1 resection<br />

R2 resection<br />

T1, N0<br />

T2, N0<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

T3, T4 or<br />

Any T, N+<br />

ADJUNCTIVE TREATMENT<br />

RT, 45–50.4 Gy + concurrent<br />

5-FU-based radiosensitization (preferred)<br />

+ 5-FU ± leucovor<strong>in</strong><br />

RT, 45–50.4 Gy + concurrent<br />

5-FU-based radiosensitization<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

or<br />

Chemotherapy<br />

or<br />

Best supportive care<br />

(poor performance status)<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

f<br />

Observe<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Follow-up (see GAST-4)<br />

Observe or Chemotherapy<br />

( 5-FU-based)/RT<br />

for selected patientsg<br />

RT, 45–50.4 Gy + concurrent<br />

5-FU-based<br />

radiosensitization (preferred)<br />

+ 5-FU ± leucovor<strong>in</strong><br />

Follow-up (see GAST-4)<br />

Salvage Therapy (see GAST-4)<br />

M1 Salvage Therapy (see GAST-4)<br />

Restag<strong>in</strong>g (preferred):<br />

� Chest x-ray<br />

Adjunctive<br />

� Abdom<strong>in</strong>al CT<br />

treatment,<br />

� Pelvic imag<strong>in</strong>g<br />

postchemo-<br />

(females)<br />

therapy ± RT<br />

� CBC, SMA-12<br />

� PET/CT scan<br />

eSee<br />

Pr<strong>in</strong>ciples of Surgery (GAST-A) .<br />

fSee<br />

Pr<strong>in</strong>ciples of Systemic Therapy (GAST-B) .<br />

Complete response<br />

or major response<br />

Residual,<br />

locoregional<br />

and/or<br />

distant metastases<br />

Follow-up<br />

(see GAST-4)<br />

or<br />

Surgery, e if<br />

appropriate<br />

Salvage Therapy (see GAST-4)<br />

gHigh risk features such as poorly differentiated or higher grade cancer, lymphovascular <strong>in</strong>vasion, neural <strong>in</strong>vasion, or < 50 years of age.<br />

f<br />

GAST-3


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

FOLLOW-UP<br />

� H&P every 4-6mofor3y,<br />

then annually<br />

� CBC, platelets, SMA-12, as<br />

<strong>in</strong>dicated<br />

� Radiologic imag<strong>in</strong>g or<br />

endoscopy, as cl<strong>in</strong>ically<br />

<strong>in</strong>dicated<br />

� Monitor vitam<strong>in</strong> B12 for<br />

proximal or total<br />

gastrectomy patientsh<br />

SALVAGE THERAPY<br />

Karnofsky performance<br />

score > 60<br />

or<br />

ECOG performance<br />

score � 2<br />

Karnofsky performance<br />

score � 60<br />

or<br />

ECOG performance<br />

score � 3<br />

fSee<br />

Pr<strong>in</strong>ciples of Systemic Therapy (GAST-B).<br />

hPatients should be monitored for vitam<strong>in</strong> B12 deficiency and treated as <strong>in</strong>dicated.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Chemotherapy<br />

or<br />

Cl<strong>in</strong>ical trial<br />

Best<br />

supportive<br />

care<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

or<br />

Best supportive<br />

care<br />

f<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Supportive Care Modalities<br />

� Obstruction: Stent, laser,<br />

photodynamic therapy, RT, surgery<br />

� Nutrition: Enteral feed<strong>in</strong>g, nutritional<br />

counsel<strong>in</strong>g<br />

� Pa<strong>in</strong> control: RT and/or medications<br />

� Bleed<strong>in</strong>g: RT, surgery or endoscopic<br />

therapy<br />

GAST-4


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Surgery<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

PRINCIPLES OF SURGERY<br />

Type:<br />

� Distal (body + antrum): prefer subtotal gastrectomy<br />

� Proximal (cardia): total or proximal gastrectomy, as <strong>in</strong>dicated<br />

� Splenectomy: avoid if possible<br />

� Consider plac<strong>in</strong>g a feed<strong>in</strong>g jejunostomy tube<br />

� Prefer >5cmproximal and distal marg<strong>in</strong>s from gross tumor<br />

Criteria for unresectability for cure:<br />

� Peritoneal seed<strong>in</strong>g or distant metastases<br />

� Inability to perform a complete resection<br />

� Invasion or encasement of major vascular structure<br />

Extent of lymph node dissection recommended:<br />

� D0: unacceptable<br />

� M<strong>in</strong>imum of 15 lymph nodes should be evaluated<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

GAST-A


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Preoperative Chemotherapy:<br />

� ECF (category 1)<br />

Preoperative Chemoradiation<br />

(Recommended <strong>in</strong> localized unresectable case) :<br />

� Fluoropyrimid<strong>in</strong>e/leucovor<strong>in</strong> (category 2B)<br />

� Fluoropyrimid<strong>in</strong>e-based (category 2B)<br />

� Cisplat<strong>in</strong>-based (category 2B)<br />

� Taxane-based (category 2B)<br />

� Ir<strong>in</strong>otecan-based (category 2B)<br />

Postoperative Chemoradiation:<br />

� Fluoropyrimid<strong>in</strong>e/leucovor<strong>in</strong> (category 1)<br />

� Fluoropyrimid<strong>in</strong>e-based (category 1)<br />

� Fluoropyrimid<strong>in</strong>e/cisplat<strong>in</strong> (category 2B)<br />

� ECF (category 2B)<br />

� Taxane-based (category 2B)<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

PRINCIPLES OF SYSTEMIC THERAPY<br />

Postoperative Chemotherapy:<br />

� ECF (Only when preoperative ECF has been adm<strong>in</strong>istered)<br />

(category 1)<br />

Metastatic <strong>Cancer</strong>:<br />

� Fluoropyrimid<strong>in</strong>e/leucovor<strong>in</strong> (category 2B)<br />

� Fluoropyrimid<strong>in</strong>e-based (category 2B)<br />

� Cisplat<strong>in</strong>-based (category 2B)<br />

� Oxaliplat<strong>in</strong>-based (category 2B)<br />

� Taxane-based (category 1)<br />

� Ir<strong>in</strong>otecan-based (category 2B)<br />

� ECF (category 1)<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

� For resected gastric carc<strong>in</strong>oma, only f luoropyrimid<strong>in</strong>e/leucovor<strong>in</strong><br />

has been studied <strong>in</strong> conjunction with radiation therapy <strong>in</strong> a phase III<br />

sett<strong>in</strong>g (Intergroup 116). 1 However, many participat<strong>in</strong>g <strong>in</strong>stitutions have developed chemotherapy variations <strong>in</strong> the context of phase II<br />

studies. Thus, many regimens <strong>in</strong>dicated below represent <strong>in</strong>stitutional preferences but they may not be superior to<br />

f luoropyrimid<strong>in</strong>e/leucovor<strong>in</strong>.<br />

� For metastatic gastric carc<strong>in</strong>oma: there have been only a few phase III trials (experimental arms be<strong>in</strong>g: ECF (Epirubic<strong>in</strong>/cisplat<strong>in</strong>/5-FU),<br />

DCF (Docetaxel/cisplat<strong>in</strong>/5-FU), and FOLFIRI (AIO regimen Infusional 5-FU/leucovor<strong>in</strong>/ir<strong>in</strong>otecan). The regimens <strong>in</strong>dicated below <strong>in</strong>clude<br />

<strong>in</strong>stitutional preferences <strong>in</strong> the context of phase II trials. The regimens not studied <strong>in</strong> the phase III sett<strong>in</strong>g may not be superior to DCF or<br />

ECF.<br />

� It should be noted that there is no established second-l<strong>in</strong>e therapy for advanced gastric cancer. Moreover, many regimens may be<br />

considered as reference regimens <strong>in</strong> the first-l<strong>in</strong>e sett<strong>in</strong>g.<br />

1Macdonald JS, Smalley SR, Benedetti J, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarc<strong>in</strong>oma of the stomach or gastroesophageal<br />

junction. N Engl J Med. Sep 6;345(10):725-30, 2001.<br />

Note: All recommendations are category 2A unless otherwise <strong>in</strong>dicated.<br />

Cl<strong>in</strong>ical Trials: NCCN believes that the best management of any cancer patient is <strong>in</strong> a cl<strong>in</strong>ical trial. Participation <strong>in</strong> cl<strong>in</strong>ical trials is especially encouraged.<br />

GAST-B


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Stag<strong>in</strong>g<br />

Table 1<br />

American Jo<strong>in</strong>t Committee on <strong>Cancer</strong> (AJCC) TNM Stag<strong>in</strong>g<br />

Classification for Carc<strong>in</strong>oma of the Stomach*<br />

Primary Tumor (T)<br />

TX Primary tumor cannot be assessed<br />

T0 No evidence of primary tumor<br />

Tis Carc<strong>in</strong>oma <strong>in</strong> situ: <strong>in</strong>traepithelial tumor without <strong>in</strong>vasion of the<br />

lam<strong>in</strong>a propria<br />

T1 Tumor <strong>in</strong>vades lam<strong>in</strong>a propria or submucosa<br />

T2 Tumor <strong>in</strong>vades muscularis propria or subserosa†<br />

T2a Tumor <strong>in</strong>vades muscularis propria<br />

T2b Tumor <strong>in</strong>vades subserosa<br />

T3 Tumor penetrates serosa (visceral peritoneum) without<br />

<strong>in</strong>vasion of adjacent structures‡<br />

T4 Tumor <strong>in</strong>vades adjacent structures‡<br />

Regional Lymph Nodes (N)<br />

NX Regional lymph node(s) cannot be assessed<br />

N0 No regional lymph node metastasis§<br />

N1 Metastasis <strong>in</strong> 1 to 6 regional lymph nodes<br />

N2 Metastasis <strong>in</strong> 7 to 15 regional lymph nodes<br />

N3 Metastasis <strong>in</strong> more than 15 regional lymph nodes<br />

Distant Metastasis (M)<br />

MX Distant metastasis cannot be assessed<br />

M0 No distant metastasis<br />

M1 Distant metastasis<br />

Histologic Grade (G)<br />

GX Grade cannot be assessed<br />

G1 Well differentiated<br />

G2 Moderately differentiated<br />

G3 Poorly differentiated<br />

G4 Undifferentiated<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Stage Group<strong>in</strong>g<br />

Stage 0 Tis N0 M0<br />

Stage IA T1 N0 M0<br />

Stage IB T1 N1 M0<br />

T2a/b N0 M0<br />

Stage II T1 N2 M0<br />

T2a/b N1 M0<br />

T3 N0 M0<br />

Stage IIIA T2a/b N2 M0<br />

T3 N1 M0<br />

T4 N0 M0<br />

Stage IIIB T3 N2 M0<br />

Stage IV T4 N1-3 M0<br />

T1-3 N3 M0<br />

Any T Any N M1<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

*Used with permission of the American Jo<strong>in</strong>t Committee on <strong>Cancer</strong><br />

(AJCC), Chicago, Ill<strong>in</strong>ois. The orig<strong>in</strong>al and primary source for this<br />

<strong>in</strong>formation is the AJCC <strong>Cancer</strong> Stag<strong>in</strong>g Manual, Sixth Edition (2002)<br />

published by Spr<strong>in</strong>ger-Verlag New York. (For more <strong>in</strong>formation, visit<br />

www.cancerstag<strong>in</strong>g.net.)<br />

Any citation or quotation of this material must be<br />

credited to the AJCC as its primary source. The <strong>in</strong>clusion of this<br />

<strong>in</strong>formation here<strong>in</strong> does not authorize any reuse or further distribution<br />

without the expressed written permission of Spr<strong>in</strong>ger-Verlag New York on<br />

behalf of the AJCC.<br />

†A tumor may penetrate the muscularis propria with extension <strong>in</strong>to the<br />

gastrocolic or gastrohepatic ligaments, or <strong>in</strong>to the greater or lesser<br />

omentum, without perforation of the visceral peritoneum cover<strong>in</strong>g these<br />

structures. In this case, the tumor is classified as T2. If there is perforation<br />

of the visceral peritoneum cover<strong>in</strong>g the gastric ligaments or the omentum,<br />

the tumor should be classified as T3.<br />

‡The adjacent structures of the stomach <strong>in</strong>clude the spleen, transverse<br />

colon, liver, diaphragm, pancreas, abdom<strong>in</strong>al wall, adrenal gland, kidney,<br />

small <strong>in</strong>test<strong>in</strong>e, and retroperitoneum. Intramural extension to the<br />

duodenum or esophagus is classified by the depth of the greatest <strong>in</strong>vasion<br />

<strong>in</strong> any of these sites, <strong>in</strong>clud<strong>in</strong>g the stomach.<br />

§A designation of pN0 should be used if all exam<strong>in</strong>ed lymph nodes are<br />

negative, regardless of the total number removed and exam<strong>in</strong>ed.<br />

ST-1


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Manuscript<br />

NCCN Categories of Consensus<br />

This manuscript is be<strong>in</strong>g updated to correspond<br />

with the newly updated algorithm.<br />

Category 1:<br />

There is uniform NCCN consensus, based on high-level<br />

evidence, that the recommendation is appropriate.<br />

Category 2A:<br />

There is uniform NCCN consensus, based on lowerlevel<br />

evidence <strong>in</strong>clud<strong>in</strong>g cl<strong>in</strong>ical experience, that the<br />

recommendation is appropriate.<br />

Category 2B:<br />

There is nonuniform NCCN consensus (but no major<br />

disagreement), based on lower-level evidence <strong>in</strong>clud<strong>in</strong>g cl<strong>in</strong>ical<br />

experience, that the recommendation is appropriate.<br />

Category 3:<br />

There is major NCCN disagreement that the<br />

recommendation is appropriate.<br />

All recommendations are category 2A unless otherwise noted.<br />

Overview<br />

Carc<strong>in</strong>omas orig<strong>in</strong>at<strong>in</strong>g <strong>in</strong> the upper gastro<strong>in</strong>test<strong>in</strong>al (GI) tract (esophagus,<br />

gastroesophageal junction, and stomach) constitute a major<br />

health problem around the world. It is estimated that approximately<br />

36,830 new cases of upper GI carc<strong>in</strong>omas and 25,200 deaths will<br />

1<br />

occur <strong>in</strong> the United States <strong>in</strong> 2006. There has been a dramatic shift <strong>in</strong><br />

2<br />

the location of upper GI tumors <strong>in</strong> the United States. Changes <strong>in</strong><br />

histology as well as location of upper GI tumors have also been<br />

3-5<br />

observed <strong>in</strong> some parts of Europe. In countries <strong>in</strong> the Western<br />

Hemisphere, gastric carc<strong>in</strong>oma has migrated proximally; it occurs most<br />

frequently along the proximal lesser curvature, <strong>in</strong> the cardia, and <strong>in</strong> the<br />

2<br />

gastroesophageal junction. It is possible that <strong>in</strong> the com<strong>in</strong>g decades<br />

these chang<strong>in</strong>g trends will also occur <strong>in</strong> South America and Asia.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Epidemiology of <strong>Gastric</strong> Carc<strong>in</strong>oma<br />

<strong>Gastric</strong> carc<strong>in</strong>oma is rampant <strong>in</strong> many countries around the world.<br />

By some estimates, it is the second most common malignant<br />

disorder worldwide. Its <strong>in</strong>cidence, however, has been decl<strong>in</strong><strong>in</strong>g<br />

globally s<strong>in</strong>ce World War II. <strong>Gastric</strong> carc<strong>in</strong>oma is one of the least<br />

common cancers <strong>in</strong> North America. Nevertheless, it rema<strong>in</strong>s the<br />

eighth lead<strong>in</strong>g cause of cancer death <strong>in</strong> the United States. In 2006,<br />

more than 22,280 new cases of gastric cancer are estimated to<br />

occur <strong>in</strong> the United States and 11,430 deaths are expected as a<br />

1<br />

result. In developed countries, the <strong>in</strong>cidence of gastric cancer<br />

localized to the cardia follows the distribution of esophageal cancer;<br />

however, unlike the latter, the rates of gastric cancer have stabilized<br />

6-8<br />

s<strong>in</strong>ce 1998. Noncardia gastric adenocarc<strong>in</strong>oma also shows<br />

marked geographic variation; thus, countries such as Japan, Costa<br />

Rica, Peru, Brazil, Ch<strong>in</strong>a, Korea, Chile, Taiwan, and the former<br />

9,10<br />

Soviet Union show a high <strong>in</strong>cidence of the cancer. In Japan,<br />

gastric cancer rema<strong>in</strong>s the most common type of cancer among<br />

men. In contrast to the <strong>in</strong>creas<strong>in</strong>g <strong>in</strong>cidence of proximal tumors <strong>in</strong><br />

the West, non-proximal tumors cont<strong>in</strong>ue to predom<strong>in</strong>ate <strong>in</strong> Japan<br />

11,12<br />

and other parts of the world. The cause of this shift rema<strong>in</strong>s<br />

elusive and may be multifactorial.<br />

<strong>Gastric</strong> carc<strong>in</strong>oma is often diagnosed at an advanced stage,<br />

because screen<strong>in</strong>g for gastric carc<strong>in</strong>oma is not performed <strong>in</strong> most of<br />

the world, except <strong>in</strong> Japan (and <strong>in</strong> a limited fashion <strong>in</strong> Korea) where<br />

early detection of gastric carc<strong>in</strong>oma is often done. Thus, gastric<br />

carc<strong>in</strong>oma cont<strong>in</strong>ues to pose a major challenge for healthcare<br />

professionals. Risk factors <strong>in</strong>clude Helicobacter pylori <strong>in</strong>fection,<br />

smok<strong>in</strong>g, high salt <strong>in</strong>take, and other dietary factors. A few gastric<br />

cancers (1%-3%) are associated with <strong>in</strong>herited gastric cancer<br />

predisposition syndromes. E-cadher<strong>in</strong> mutations occur <strong>in</strong> an<br />

MS-1


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

estimated 25% of families with an autosomal dom<strong>in</strong>ant<br />

predisposition to diffuse type gastric cancers; this subset of gastric<br />

cancer has been termed hereditary diffuse gastric cancer. Data<br />

suggest it may be useful to provide genetic counsel<strong>in</strong>g and to<br />

consider prophylactic gastrectomy <strong>in</strong> young, asymptomatic carriers<br />

of germ-l<strong>in</strong>e truncat<strong>in</strong>g CDH1 mutations who belong to families with<br />

highly penetrant hereditary diffuse gastric cancer.<br />

13<br />

14<br />

Stag<strong>in</strong>g<br />

Two major classification systems are currently <strong>in</strong> use for gastric<br />

carc<strong>in</strong>oma. The most elaborate of these, the Japanese<br />

classification, is based on ref<strong>in</strong>ed anatomic <strong>in</strong>volvement, particularly<br />

15<br />

the lymph node stations. The other stag<strong>in</strong>g system for gastric<br />

carc<strong>in</strong>oma, developed jo<strong>in</strong>tly by the American Jo<strong>in</strong>t Committee on<br />

<strong>Cancer</strong> (AJCC) and the International Union Aga<strong>in</strong>st <strong>Cancer</strong> (UICC),<br />

is based on a gastric cancer database and demonstrates that the<br />

prognosis of node-positive patients depends on the number of lymph<br />

16<br />

nodes <strong>in</strong>volved. The modern stag<strong>in</strong>g of gastric carc<strong>in</strong>oma is based<br />

on this tumor/node/metastasis (TNM) classification, rather than on<br />

the size of the cancer. The AJCC/UICC classification (see Table 1)<br />

is<br />

the system used <strong>in</strong> countries <strong>in</strong> the Western Hemisphere.<br />

Patient outcome depends on the <strong>in</strong>itial stage of the cancer at<br />

diagnosis. However, at diagnosis, approximately 50% of patients<br />

have gastric carc<strong>in</strong>oma that extends beyond the locoregional<br />

conf<strong>in</strong>es. In addition, approximately 50% of patients with<br />

locoregional gastric carc<strong>in</strong>oma cannot undergo a curative resection<br />

17,18<br />

(R0). Note that the R classification refers to the amount of<br />

residual cancer rema<strong>in</strong><strong>in</strong>g after tumor resection: R0 <strong>in</strong>dicates no<br />

macroscopic or microscopic cancer at resection marg<strong>in</strong>s (ie,<br />

negative marg<strong>in</strong>s); R1 <strong>in</strong>dicates microscopic residual cancer (ie,<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update Surgery<strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

positive marg<strong>in</strong>s); and R2 <strong>in</strong>dicates gross (macroscopic) residual<br />

19<br />

cancer (ie, positive marg<strong>in</strong>s) but not distant disease. Although<br />

surgical pathology yields the most accurate stage, cl<strong>in</strong>ical stag<strong>in</strong>g<br />

has been greatly improved by advancements <strong>in</strong> imag<strong>in</strong>g techniques,<br />

<strong>in</strong>clud<strong>in</strong>g laparoscopic evaluation of the peritoneal cavity and liver<br />

as well as endoscopic ultrasonography to assess the primary tumor<br />

20<br />

and regional lymph nodes. Nearly 70% to 80% of resected gastric<br />

carc<strong>in</strong>oma specimens have metastases <strong>in</strong> the regional lymph nodes.<br />

Thus, it is common to encounter patients with advanced gastric<br />

carc<strong>in</strong>oma at presentation. Poor prognostic factors <strong>in</strong> patients with<br />

locally advanced and metastatic esophago-gastric cancer <strong>in</strong>clude:<br />

poor performance status (2 or more), liver metastases, peritoneal<br />

21<br />

metastases, and alkal<strong>in</strong>e phosphatase of 100 U/L or more.<br />

Surgical therapy is the primary treatment for gastric carc<strong>in</strong>oma.<br />

Widely agreed on surgical pr<strong>in</strong>ciples for the management of gastric<br />

cancer <strong>in</strong>clude complete resection with adequate marg<strong>in</strong>s (5 cm).<br />

The type of resection (subtotal versus total gastrectomy) and the<br />

role of extensive lymphadenectomy have been the subjects of<br />

<strong>in</strong>ternational debate.<br />

For distal gastric cancers, subtotal gastrectomy has been shown to<br />

have an equivalent oncologic result with significantly fewer<br />

22<br />

complications when compared with total gastrectomy. The surgical<br />

procedure of choice for proximal gastric cancers is more<br />

controversial, because both procedures (proximal gastrectomy and<br />

total gastrectomy) are associated with postoperative nutritional<br />

impairments. Currently, most authorities advocate total gastrectomy<br />

for proximal (cardia) tumors.<br />

MS-2


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Even more controversial is the extent of lymphatic dissection that is<br />

required. The Japanese Research Society for the Study of <strong>Gastric</strong><br />

<strong>Cancer</strong> has established guidel<strong>in</strong>es for pathologic exam<strong>in</strong>ation and<br />

12<br />

evaluation of lymph node stations that surround the stomach. The<br />

perigastric lymph node stations along the lesser curvature (stations<br />

1, 3, and 5) and greater curvature (stations 2, 4, and 6) of the<br />

stomach are grouped together as N1. The nodes along the left<br />

gastric artery (station 7), common hepatic artery (station 8), celiac<br />

artery (station 9), and splenic artery (stations 10 and 11) are<br />

grouped together as N2. More distant nodes, <strong>in</strong>clud<strong>in</strong>g para-aortic<br />

(N3 and N4), are regarded as distant metastases.<br />

A D1 dissection entails the removal of the <strong>in</strong>volved distal part of the<br />

stomach or the entire stomach (distal or total resection), <strong>in</strong>clud<strong>in</strong>g<br />

the greater and lesser omenta. For a D2 dissection, the omental<br />

bursa is removed, along with the front leaf of the transverse<br />

mesocolon, and the mentioned arteries are cleared completely. A<br />

splenectomy (to remove stations 10 and 11) is required for a D2<br />

dissection for proximal gastric tumors. If N1 lymph nodes are not<br />

removed, then this is def<strong>in</strong>ed as a D0 dissection. The technical<br />

aspects of perform<strong>in</strong>g a D2 dissection require a significant degree of<br />

tra<strong>in</strong><strong>in</strong>g and expertise. In an Intergroup trial exam<strong>in</strong><strong>in</strong>g the role of<br />

adjuvant therapy for gastric cancer, 54% of the patients had a D0<br />

lymphadenectomy, whereas only 10% of patients had the<br />

23<br />

recommended D2 lymphadenectomy.<br />

Japanese <strong>in</strong>vestigators have often emphasized the value of<br />

extensive lymphadenectomy (D2 and above); however, Western<br />

<strong>in</strong>vestigators have not found a survival advantage when extensive<br />

24<br />

lymphadenectomy is compared with a D1 resection. The Dutch<br />

<strong>Gastric</strong> <strong>Cancer</strong> Group Trial recently published long-term survival<br />

25<br />

data compar<strong>in</strong>g D1 versus D2 resection. A total of 711 patients who<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

underwent surgical resection with curative <strong>in</strong>tent were randomly<br />

assigned to either a D1 or D2 lymphadenectomy. When compared<br />

with the D1 dissection, both the morbidity (25% versus 43%, P <<br />

.001) and mortality (4% versus 10%, P = .004) were higher for the<br />

D2 dissection, with no difference <strong>in</strong> overall survival (30% versus<br />

35%, P = .53). The authors identified splenectomy, pancreatectomy,<br />

and age older than 70 years as contribut<strong>in</strong>g risk factors for<br />

<strong>in</strong>creased morbidity and mortality. In a subset analysis, a trend to<br />

improved survival appeared to occur <strong>in</strong> patients with N2 cancer<br />

undergo<strong>in</strong>g a D2 lymphadenectomy. Unfortunately, N2 cancer can<br />

only be detected after microscopic exam<strong>in</strong>ation of the surgical<br />

specimen. A similar study conducted by the Medical Research<br />

Council failed to demonstrate a survival benefit of D2 over D1<br />

26<br />

lymphadenectomy. In addition, the D2 dissection was associated<br />

with <strong>in</strong>creased morbidity and mortality. A meta-analysis did not show<br />

any survival benefit from extended lymph node dissections but did<br />

27<br />

show <strong>in</strong>creased mortality.<br />

Despite these results, <strong>in</strong>terest <strong>in</strong> extended lymphatic dissections (D2<br />

28<br />

and greater) has not waned. Investigators have argued that if the<br />

complication rate after a D2 operation could be decreased then<br />

there may be a benefit <strong>in</strong> selected patients. A recent phase II study<br />

of D2 dissection by the Italian <strong>Gastric</strong> <strong>Cancer</strong> Study Group (IGCSG)<br />

has demonstrated a morbidity of 20.9% and a postoperative<br />

29<br />

mortality rate of 3%. These rates are comparable to the rates for<br />

D1 dissections <strong>in</strong> the Dutch and United K<strong>in</strong>gdom trial. The difference<br />

<strong>in</strong> the IGCSG trial was the lack of rout<strong>in</strong>e pancreatectomy <strong>in</strong> patients<br />

with proximal gastric tumors (except when warranted for direct<br />

<strong>in</strong>vasion). Japanese <strong>in</strong>vestigators compar<strong>in</strong>g D2 versus extended<br />

D2 (<strong>in</strong>clud<strong>in</strong>g para-aortic lymph nodes) have recently reported a<br />

30<br />

postoperative morality rate of 0.8% <strong>in</strong> each arm. Survival data from<br />

MS-3


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

this study are currently not available. A surgical option that may<br />

decrease morbidity and mortality is an “over-D1” (ie, D1+)<br />

lymphadenectomy with preservation of the pancreatic tail and<br />

31, 32<br />

without splenectomy.<br />

With improvements <strong>in</strong> endoscopic techniques (endoscopic mucosal<br />

resection [EMR]) and m<strong>in</strong>imal access surgery (laparoscopic wedge<br />

resection), there has been <strong>in</strong>terest <strong>in</strong> apply<strong>in</strong>g these modalities to<br />

early gastric cancer (T1, mucosal and submucosal). Node-negative<br />

33<br />

T1 tumors are associated with a 5-year survival of more than 90%.<br />

As such, there is <strong>in</strong>terest <strong>in</strong> perform<strong>in</strong>g more limited resection for<br />

these tumors. Proper patient selection is paramount when<br />

employ<strong>in</strong>g endoscopic or limited gastric resections (wedge). The<br />

probability of lymph node metastasis <strong>in</strong> early gastric cancer is<br />

<strong>in</strong>fluenced by tumor factors and is <strong>in</strong>creased with <strong>in</strong>creas<strong>in</strong>g tumor<br />

size, submucosal <strong>in</strong>vasion, poorly differentiated tumors, and<br />

34<br />

lymphatic and vascular <strong>in</strong>vasion. Indications for EMR <strong>in</strong>clude welldifferentiated<br />

or moderately differentiated histology, tumor size less<br />

than 30 mm, absence of ulceration, and no evidence of <strong>in</strong>vasive<br />

35<br />

f<strong>in</strong>d<strong>in</strong>gs. Regardless of the technique used for resect<strong>in</strong>g early<br />

gastric tumors, complete excision with negative marg<strong>in</strong>s is required.<br />

Endoscopic ultrasound may be useful <strong>in</strong> assess<strong>in</strong>g the depth of<br />

36,37<br />

tumor <strong>in</strong>vasion and may aid <strong>in</strong> appropriate patient selection. Most<br />

of the experience with EMR for early gastric cancer has been ga<strong>in</strong>ed<br />

by countries with a high <strong>in</strong>cidence of gastric cancer and an active<br />

38<br />

screen<strong>in</strong>g program. The applicability of these techniques <strong>in</strong> the<br />

United States is limited because of the low <strong>in</strong>cidence of early gastric<br />

cancer. Furthermore, long-term follow-up and survival data are<br />

lack<strong>in</strong>g therefore, the rout<strong>in</strong>e use of endoscopic techniques is not<br />

recommended outside a cl<strong>in</strong>ical trial and should be limited to<br />

medical centers with extensive experience.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Radiotherapy and Chemoradiation<br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Locally Unresectable <strong>Cancer</strong><br />

Moderate-dose external-beam radiation (45-50.4 Gy) as a s<strong>in</strong>gle<br />

modality has m<strong>in</strong>imal value <strong>in</strong> palliat<strong>in</strong>g locally unresectable gastric<br />

39<br />

carc<strong>in</strong>oma and does not improve survival. However, when used<br />

concurrently with 5-fluorouracil (5-FU), moderate-dose external-beam<br />

40<br />

radiation does improve survival. Moertel and colleagues assessed<br />

5-FU plus 35 to 40 Gy of radiotherapy compared with radiotherapy<br />

alone <strong>in</strong> the treatment of locally unresectable gastric carc<strong>in</strong>oma. They<br />

observed a 6-month survival advantage <strong>in</strong> the group receiv<strong>in</strong>g comb<strong>in</strong>ed<br />

modality therapy. In another study by the Gastro<strong>in</strong>test<strong>in</strong>al<br />

Tumor Study Group, 90 patients with locally advanced gastric carc<strong>in</strong>oma<br />

were randomly assigned to receive either comb<strong>in</strong>ation chemotherapy<br />

(5-FU plus methyl-CCNU [lomust<strong>in</strong>e]) or split-course radiation<br />

therapy (RT) with a concurrent <strong>in</strong>travenous bolus of 5-FU given<br />

dur<strong>in</strong>g the first 3 days of 2 sessions of 25 Gy, separated by a 2-week<br />

41<br />

break, and followed by ma<strong>in</strong>tenance 5-FU plus methyl-CCNU. In the<br />

first 26 weeks, mortality was higher <strong>in</strong> the comb<strong>in</strong>ed modality group.<br />

At 3 years, however, the survival curve reached a plateau <strong>in</strong> the<br />

comb<strong>in</strong>ed modality arm, but tumor-related deaths cont<strong>in</strong>ued to occur<br />

<strong>in</strong> the chemotherapy-alone arm, suggest<strong>in</strong>g that a small fraction of<br />

patients can be cured with comb<strong>in</strong>ed modality therapy.<br />

This approach needs to be further developed <strong>in</strong> light of newly<br />

available radioenhancers. New agents---such as taxanes,<br />

epirubic<strong>in</strong>, and ir<strong>in</strong>otecan---have been used <strong>in</strong> comb<strong>in</strong>ation with<br />

42-44<br />

RT. Results of the comparative trials are pend<strong>in</strong>g.<br />

Preoperative or Postoperative Chemotherapy<br />

Recent studies suggest that preoperative <strong>in</strong>duction chemotherapy<br />

followed by chemoradiotherapy yields a substantial pathologic<br />

MS-4


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

45,46<br />

response that results <strong>in</strong> durable survival time. However, the value<br />

of such approaches needs to be determ<strong>in</strong>ed <strong>in</strong> comparative trials.<br />

47<br />

Nonrandomized trials from Baeza and colleagues have reported<br />

encourag<strong>in</strong>g results for patients with R0 resections who receive<br />

adjunctive treatment. Limited reports from randomized trials of<br />

postoperative RT with or without chemotherapy after a complete<br />

resection with negative marg<strong>in</strong>s did not reveal a clear survival<br />

48,49<br />

advantage.<br />

23,50<br />

The landmark trial is the Intergroup trial INT-0116. Eligibility<br />

<strong>in</strong>cluded patients with T3 and/or N+ adenocarc<strong>in</strong>oma of the stomach<br />

or gastroesophageal junction. After a resection with negative<br />

marg<strong>in</strong>s, 603 patients were randomly assigned to either observation<br />

alone or postoperative comb<strong>in</strong>ed modality therapy consist<strong>in</strong>g of 5<br />

monthly cycles of bolus chemotherapy with 45 Gy concurrent with<br />

cycles 2 and 3. There was a significant decrease <strong>in</strong> local failure as<br />

the first site of failure (19% versus 29%) as well as an <strong>in</strong>crease <strong>in</strong><br />

median survival (36 versus 27 months), 3-year relapse-free survival<br />

(48% versus 31%), and overall survival (50% versus 41%, P = .005)<br />

with comb<strong>in</strong>ed modality therapy. The CALGB 80101 phase III trial is<br />

currently assess<strong>in</strong>g postoperative standard therapy with 5-<br />

FU/leucovor<strong>in</strong>/radiation versus ECF (epirubic<strong>in</strong>, cisplat<strong>in</strong>, and 5-<br />

FU)/radiation<br />

( http://www.nci.nih.gov/search/ViewCl<strong>in</strong>icalTrials.aspx?cdrid=25878<br />

7&version=patient&protocolsearchid=1575831).<br />

51<br />

Smalley and colleagues reviewed gastric anatomy and patterns of<br />

failure after surgery, and they offer detailed radiation treatment<br />

plann<strong>in</strong>g recommendations. A randomized trial by Zhang and<br />

associates from Beij<strong>in</strong>g revealed a significant improvement <strong>in</strong><br />

52<br />

survival with preoperative radiation (30% versus 20%, P = .0094).<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

These data suggest that preoperative radiation improves local<br />

control and survival. However, randomized trials are needed to<br />

confirm these results <strong>in</strong> patients from the Western Hemisphere.<br />

The sem<strong>in</strong>al trial exam<strong>in</strong><strong>in</strong>g the role of postoperative comb<strong>in</strong>ed<br />

modality therapy <strong>in</strong> gastric cancer was reported by Moertel and<br />

40<br />

colleagues <strong>in</strong> 1969 (40 Gy versus 40 Gy plus 5-FU). This trial<br />

revealed a significant improvement <strong>in</strong> survival. The rema<strong>in</strong><strong>in</strong>g<br />

randomized trials <strong>in</strong>clude patients with unresectable or residual<br />

cancer. None have shown a survival advantage. The use of<br />

<strong>in</strong>traoperative RT rema<strong>in</strong>s <strong>in</strong>vestigational.<br />

For resected gastric carc<strong>in</strong>oma, only 5-FU/leucovor<strong>in</strong> (category 1)<br />

has been studied <strong>in</strong> conjunction with RT <strong>in</strong> a phase III sett<strong>in</strong>g<br />

23<br />

(Intergroup 116). However, many participat<strong>in</strong>g <strong>in</strong>stitutions have<br />

developed other chemotherapy regimens <strong>in</strong> the context of phase II<br />

studies. Thus, these regimens represent <strong>in</strong>stitutional preferences,<br />

but they may not be superior to 5-FU/leucovor<strong>in</strong>. In the NCCN<br />

algorithm, preoperative chemoradiation options for localized,<br />

unresectable disease <strong>in</strong>clude 5-FU/leucovor<strong>in</strong> (category 1) as well<br />

as the follow<strong>in</strong>g category 3 options such as 5-FU--based, cisplat<strong>in</strong>based,<br />

taxane-based, and ir<strong>in</strong>otecan-based regimens. Postoperative<br />

chemoradiation options <strong>in</strong>clude 5-FU/leucovor<strong>in</strong> (category 1) as well<br />

as the follow<strong>in</strong>g category 3 options such as 5-FU/cisplat<strong>in</strong>, 5-FUbased,<br />

taxane-based, and ECF regimens.<br />

Chemotherapy<br />

Advanced gastric carc<strong>in</strong>oma is <strong>in</strong>curable, but chemotherapy can<br />

have a palliative effect <strong>in</strong> symptomatic patients. In four studies,<br />

comb<strong>in</strong>ation chemotherapy resulted <strong>in</strong> better quality of life and<br />

overall survival when compared with best supportive care <strong>in</strong> patients<br />

53-56<br />

with advanced gastric carc<strong>in</strong>oma. However, all four studies only<br />

MS-5


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

had a small number of patients. Only a few s<strong>in</strong>gle agents have<br />

established activity aga<strong>in</strong>st advanced gastric carc<strong>in</strong>oma; these<br />

57 58<br />

agents <strong>in</strong>clude 5-FU, mitomyc<strong>in</strong>, etoposide, and cisplat<strong>in</strong>.<br />

In the early 1980s, the FAM (5-FU, doxorubic<strong>in</strong>, and mitomyc<strong>in</strong>)<br />

regimen was the gold standard therapy for patients with advanced<br />

59<br />

gastric carc<strong>in</strong>oma. In a pivotal study performed by the North<br />

60<br />

Central <strong>Cancer</strong> Treatment Group (NCCTG), the FAM regimen was<br />

compared with 5-FU as a s<strong>in</strong>gle agent and 5-FU plus doxorubic<strong>in</strong>.<br />

No significant survival difference was detected among patients<br />

treated with these three regimens. However, response rates were<br />

higher with comb<strong>in</strong>ation chemotherapy than with 5-FU alone. Thus,<br />

comb<strong>in</strong>ation chemotherapy is preferable to s<strong>in</strong>gle-agent therapy for<br />

palliation.<br />

Several randomized studies compar<strong>in</strong>g FAM versus FAMTX (5-FU,<br />

61<br />

adriamyc<strong>in</strong>, and methotrexate [with leucovor<strong>in</strong> rescue]), FAMTX<br />

62<br />

versus ECF, and FAMTX versus ELF (etoposide, leucovor<strong>in</strong>, and 5-<br />

63<br />

FU) versus 5-FU plus cisplat<strong>in</strong> have been reported <strong>in</strong> the past<br />

several years. No one standard therapy has emerged from these<br />

trials. Outside of cl<strong>in</strong>ical trials, the recommended chemotherapy for<br />

advanced gastric carc<strong>in</strong>oma is either cisplat<strong>in</strong>-based or 5-FU--based<br />

comb<strong>in</strong>ation chemotherapy.<br />

Several drugs and their comb<strong>in</strong>ations have shown activity aga<strong>in</strong>st<br />

64-66 67-69<br />

gastric carc<strong>in</strong>oma. The agents <strong>in</strong>clude paclitaxel, docetaxel,<br />

70 71<br />

ir<strong>in</strong>otecan, UFT (a comb<strong>in</strong>ation of uracil and tegafur), oral<br />

72 73-77<br />

etoposide, and S-1. In addition, comb<strong>in</strong>ation chemotherapy<br />

78-89<br />

regimens have also been assessed. A number of oral agents also<br />

81,82<br />

hold promise <strong>in</strong> the treatment of gastric carc<strong>in</strong>oma. Agents that<br />

have not been extensively studied <strong>in</strong>clude capecitab<strong>in</strong>e, oxaliplat<strong>in</strong>,<br />

and rubitecan. In addition, several new categories of agents are of<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

<strong>in</strong>terest, <strong>in</strong>clud<strong>in</strong>g vacc<strong>in</strong>es, antireceptor agents, and antiangiogenic<br />

agents. A number of chemotherapy comb<strong>in</strong>ations are currently <strong>in</strong><br />

90,91<br />

phase III trials, and we anticipate that a widely accepted front-l<strong>in</strong>e<br />

standard for patients with advanced gastric carc<strong>in</strong>oma might emerge<br />

92<br />

<strong>in</strong> the near future. For metastatic gastric carc<strong>in</strong>oma, there have<br />

been only a few phase III trials, which have assessed ECF, DCF<br />

(docetaxel/cisplat<strong>in</strong>/5-FU), and FOLFIRI-AIO (<strong>in</strong>fusional 5-<br />

FU/leucovor<strong>in</strong>/ir<strong>in</strong>otecan). However, participat<strong>in</strong>g <strong>in</strong>stitutions have<br />

developed chemotherapy regimens <strong>in</strong> the context of phase II studies<br />

. The regimens that have not been studied <strong>in</strong> the phase III sett<strong>in</strong>g<br />

may not be superior to DCF or ECF. In the NCCN algorithm, options<br />

for metastatic cancer <strong>in</strong>clude 5-FU/leucovor<strong>in</strong> (category 1) as well<br />

as the follow<strong>in</strong>g category 3 options such as 5-FU--based<br />

(capecitab<strong>in</strong>e), cisplat<strong>in</strong>-based, oxaliplat<strong>in</strong>-based, taxane-based,<br />

ir<strong>in</strong>otecan-based, and ECF regimens. Moreover, many regimens<br />

may be considered as reference regimens <strong>in</strong> the first-l<strong>in</strong>e sett<strong>in</strong>g.<br />

There is no established second-l<strong>in</strong>e therapy for advanced gastric<br />

cancer.<br />

Workup<br />

In patients with gastric cancer, present<strong>in</strong>g symptoms can <strong>in</strong>clude<br />

anemia, early satiety, weight loss, and/or bleed<strong>in</strong>g. Newly diagnosed<br />

patients should undergo a complete history, physical exam<strong>in</strong>ation,<br />

chest x-ray, and endoscopy of the entire upper GI tract. A complete<br />

blood count (CBC), platelets, multichannel serum chemistry analysis<br />

(ie, SMA-12), coagulation studies, and a computed tomography (CT)<br />

scan of the abdomen should be performed; a positron emission<br />

tomography (PET) scan may also be useful, although there may be<br />

93<br />

false-positive results with PET. Comb<strong>in</strong>ed PET/CT imag<strong>in</strong>g is more<br />

94<br />

useful than either imag<strong>in</strong>g alone for preoperative stag<strong>in</strong>g. In<br />

MS-6


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

women, a pelvic CT scan or ultrasound is also recommended.<br />

The workup permits classification of patients <strong>in</strong>to 1 of 2 groups: (1)<br />

patients with apparent locoregional carc<strong>in</strong>oma (stages I to III or M0),<br />

and (2) those with obvious metastatic carc<strong>in</strong>oma (stage IV or M1).<br />

Patients with apparent locoregional cancer can be further classified:<br />

(1) those who are medically fit (ie, able to tolerate major abdom<strong>in</strong>al<br />

surgery) and whose cancer is potentially resectable, (2) those who<br />

are medically fit but whose cancer is unresectable, and (3) those<br />

who are medically unfit.<br />

Additional Evaluation<br />

For the group of medically fit patients with apparent locoregional<br />

carc<strong>in</strong>oma, the guidel<strong>in</strong>es address the role of laparoscopy before<br />

def<strong>in</strong>itive surgery or comb<strong>in</strong>ed chemotherapy and radiation. The use of<br />

this stag<strong>in</strong>g procedure differs among the NCCN <strong>in</strong>stitutions, with<br />

several centers preferr<strong>in</strong>g laparoscopic stag<strong>in</strong>g of the peritoneal cavity<br />

for medically fit patients, whether <strong>in</strong> the potentially resectable or<br />

unresectable category (category 2B). For medically unfit patients with<br />

apparent locoregional carc<strong>in</strong>oma, laparoscopic stag<strong>in</strong>g of the peritoneal<br />

cavity can be done when consider<strong>in</strong>g chemotherapy/RT or surgery. If a<br />

palliative resection is planned, laparoscopy is not <strong>in</strong>dicated.<br />

Postlaparoscopic Stag<strong>in</strong>g<br />

If a laparoscopic exam<strong>in</strong>ation is performed, there are two<br />

possibilities for both medically fit and unfit patients with apparent<br />

locoregional carc<strong>in</strong>oma. Patients will either have apparent<br />

locoregional carc<strong>in</strong>oma or will have metastatic carc<strong>in</strong>oma (M1).<br />

Primary Therapy<br />

Surgery is recommended for medically fit patients with a potentially<br />

resectable (stages I to III) carc<strong>in</strong>oma. Medically fit patients<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

discovered to have an M1 carc<strong>in</strong>oma after laparoscopy may be<br />

offered salvage therapy. The goal of surgery is to accomplish a<br />

curative resection (R0) with negative marg<strong>in</strong>s, and 5 cm or greater<br />

proximal and distal marg<strong>in</strong>s are desirable. A D0 lymphadenectomy is<br />

unacceptable. It is recommended that at least 15 lymph nodes be<br />

removed and exam<strong>in</strong>ed. For carc<strong>in</strong>omas located <strong>in</strong> the distal<br />

stomach (body and antrum), a subtotal gastrectomy is preferred. For<br />

carc<strong>in</strong>omas located proximally (<strong>in</strong> the cardia), total gastrectomy is<br />

recommended; however, proximal gastrectomy may also be<br />

appropriate. Splenectomy should be avoided, if possible. Placement<br />

of a jejunostomy feed<strong>in</strong>g tube should be considered.<br />

Carc<strong>in</strong>omas are unresectable if there is evidence of peritoneal<br />

<strong>in</strong>volvement, distant metastases, or <strong>in</strong>vasion or encasement of<br />

major blood vessels. For medically fit patients found to have an<br />

unresectable locoregional cancer, the recommended therapy<br />

(category 1) is comb<strong>in</strong>ed RT (45 to 50.4 Gy) with concurrent 5-FU--<br />

40,41<br />

based radiosensitization. Medically unfit patients with<br />

locoregional carc<strong>in</strong>oma may be offered one of the follow<strong>in</strong>g choices:<br />

(1) RT (45 to 50.4 Gy) with concurrent 5-FU--based<br />

40,41<br />

radiosensitization (category 1); or (2) salvage chemotherapy with<br />

5-FU/leucovor<strong>in</strong> (category 1) or other agents which are category 3<br />

(such as ECF, 5-FU--based [capecitab<strong>in</strong>e], cisplat<strong>in</strong>-based,<br />

oxaliplat<strong>in</strong>-based, taxane-based, or ir<strong>in</strong>otecan-based<br />

chemotherapy). Medically unfit patients discovered to have M1<br />

carc<strong>in</strong>oma after laparoscopy may also be offered salvage therapy.<br />

Adjunctive Therapy<br />

As previously discussed, select patients with negative marg<strong>in</strong>s (R0<br />

resection) and no evidence of metastatic carc<strong>in</strong>oma after<br />

gastrectomy may receive adjuvant chemoradiation based on the<br />

MS-7


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

results of the Intergroup trial (INT-0116). However, a patient whose<br />

surgical pathologic stage is T1, N0, M0 may be observed and not<br />

treated with adjuvant therapy. All patients with an R0 resection who<br />

have T2, N0 along with high-risk features (ie, poorly differentiated or<br />

higher grade cancer, lymphovascular <strong>in</strong>vasion, neural <strong>in</strong>vasion, or<br />

age younger than 50 years) should receive adjuvant<br />

chemoradiotherapy (5-FU--based/RT); those patients without highrisk<br />

features may be observed. The panel recommends that all<br />

patients with an R0 resection who have T3, T4, or any T, N+ cancer<br />

should be offered radiotherapy (45 to 50.4 Gy) plus concurrent<br />

5-FU--based radiosensitization (preferred) plus 5-FU with or without<br />

23,40<br />

leucovor<strong>in</strong>. It should also be noted that 20% of patients <strong>in</strong> the<br />

Intergroup-0116 trial had cancers that <strong>in</strong>volved the<br />

gastroesophageal junction; therefore, adjuvant chemoradiotherapy<br />

should also be recommended for patients with similar cancers<br />

(aga<strong>in</strong>, patients with T1, N0, M0 tumors may be observed as can<br />

patients with T2, N0 without high-risk features).<br />

Patients with R1 resections should be offered radiotherapy (45 to<br />

50.4 Gy) plus concurrent 5-FU--based radiosensitization (preferred)<br />

plus 5-FU with or without leucovor<strong>in</strong>. In the absence of M1<br />

carc<strong>in</strong>oma, patients with R2 resections may be offered (1) RT (45 to<br />

50.4 Gy) with concurrent 5-FU--based radiosensitization;<br />

(2) salvage chemotherapy; or (3) best supportive care, if<br />

performance status is poor. Medically unfit patients should undergo<br />

restag<strong>in</strong>g (<strong>in</strong>clud<strong>in</strong>g chest x-ray, abdom<strong>in</strong>al CT, CBC, SMA-12,<br />

pelvic imag<strong>in</strong>g [women], PET/CT scan) after completion of<br />

95<br />

chemoradiotherapy. If a complete response of the carc<strong>in</strong>oma is<br />

determ<strong>in</strong>ed, these patients should be observed or have surgery if it<br />

is deemed appropriate. If there is evidence of residual or M1 cancer,<br />

patients may be offered salvage therapy.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Follow-up and Surveillance<br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

All patients should be followed up systematically. This follow-up<br />

should <strong>in</strong>clude a complete history and physical exam<strong>in</strong>ation every 4<br />

to 6 months for 3 years, then annually thereafter. Complete blood<br />

count, platelets, SMA-12 tests, and other <strong>in</strong>vestigations (such as<br />

endoscopy and other radiologic studies) should be done if cl<strong>in</strong>ically<br />

96<br />

<strong>in</strong>dicated. Vitam<strong>in</strong> B12 levels should be monitored for patients who<br />

have had proximal or total gastrectomy.<br />

Salvage Therapy<br />

Salvage therapy consists of either best supportive care,<br />

chemotherapy, or cl<strong>in</strong>ical trial depend<strong>in</strong>g on the patient's<br />

performance scores on the Karnofsky or Eastern Cooperative Group<br />

(ECOG) scales. The constituents of best supportive care depend on<br />

the patient's symptoms. In the case of lum<strong>in</strong>al obstruction, a patient<br />

may be offered a stent placement, laser surgery, photodynamic<br />

therapy, radiotherapy, surgery, or a comb<strong>in</strong>ation of these methods,<br />

as appropriate. For patients requir<strong>in</strong>g nutritional support, placement<br />

of a percutaneous endoscopic gastronomy (PEG) tube may be<br />

97<br />

warranted; nutritional counsel<strong>in</strong>g may also be valuable. Pa<strong>in</strong><br />

control may be achieved with the use of radiotherapy plus pa<strong>in</strong><br />

medications. Similarly, surgery, endoscopic therapy, or radiotherapy<br />

may be <strong>in</strong>dicated <strong>in</strong> patients with brisk bleed<strong>in</strong>g from the carc<strong>in</strong>oma.<br />

Whenever possible, patients should be enrolled <strong>in</strong> cl<strong>in</strong>ical trials.<br />

Outside of a cl<strong>in</strong>ical trial, patients may be treated with 5-<br />

FU/leucovor<strong>in</strong> (category 1) or other agents, which are category 3<br />

(such as ECF, 5-FU—based [capecitab<strong>in</strong>e], cisplat<strong>in</strong>-based,<br />

oxaliplat<strong>in</strong>-based, taxane-based, or ir<strong>in</strong>otecan-based<br />

chemotherapy). The decision of whether to offer best supportive<br />

care alone or with chemotherapy should be based on the patient's<br />

MS-8


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

performance status. Patients should be offered only best supportive<br />

care if they have a Karnofsky performance score of 60 or less, or an<br />

ECOG (Eastern Cooperative <strong>Oncology</strong> Group) performance score of<br />

3 or greater. Patients with a better performance status may be<br />

offered best supportive care alone, chemotherapy, or a cl<strong>in</strong>ical trial.<br />

Summary<br />

<strong>Gastric</strong> cancer is rampant <strong>in</strong> several countries around the world. Its<br />

<strong>in</strong>cidence <strong>in</strong> the Western Hemisphere has been on the decl<strong>in</strong>e for<br />

more than 40 years; however, the location of gastric cancer has<br />

shifted proximally <strong>in</strong> the past 15 years. The reason for this shift is<br />

not clear. Diffuse histology is also more common now than <strong>in</strong>test<strong>in</strong>al<br />

type of histology. Advances have been made <strong>in</strong> stag<strong>in</strong>g procedures,<br />

such as laparoscopy and endoscopic ultrasonography, and <strong>in</strong><br />

possible functional imag<strong>in</strong>g techniques. The current TNM<br />

classification requires an exam<strong>in</strong>ation of at least 15 lymph nodes; a<br />

D0 dissection is unacceptable. Patients with locoregional gastric<br />

carc<strong>in</strong>oma should also be referred to high-volume treatment centers.<br />

Comb<strong>in</strong>ation chemotherapy and radiotherapy <strong>in</strong> the adjuvant sett<strong>in</strong>g<br />

for a select group of patients is the new standard <strong>in</strong> the United<br />

States.<br />

The NCCN <strong>Gastric</strong> <strong>Cancer</strong> <strong>Guidel<strong>in</strong>es</strong> provide a uniform systematic<br />

approach to gastric cancer <strong>in</strong> the United States. We look forward to<br />

the results of <strong>in</strong>vestigations of new chemotherapeutic agents,<br />

<strong>in</strong>clud<strong>in</strong>g antireceptor agents, vacc<strong>in</strong>es, gene therapy, and<br />

antiangiogenic agents. The panel anticipates many advances <strong>in</strong> the<br />

treatment of gastric carc<strong>in</strong>oma <strong>in</strong> the future.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Disclosures for the NCCN <strong>Gastric</strong> <strong>Cancer</strong> <strong>Guidel<strong>in</strong>es</strong><br />

Panel<br />

At the beg<strong>in</strong>n<strong>in</strong>g of each panel meet<strong>in</strong>g to develop NCCN<br />

guidel<strong>in</strong>es, panel members disclosed the names of companies,<br />

foundations, and/or fund<strong>in</strong>g agencies from which they received<br />

research support; for which they participate <strong>in</strong> speakers' bureau,<br />

advisory boards; and/or <strong>in</strong> which they have equity <strong>in</strong>terest or<br />

patents. Members of the panel <strong>in</strong>dicated that they have received<br />

support from the follow<strong>in</strong>g: AstraZeneca; Berlex; Bristol Myers-<br />

Squibb; Discovery Laboratories, Inc; Exelixis; Genentech Inc;<br />

ImClone; Introgen Therapeutics, Inc; National <strong>Cancer</strong> Institute; OSI<br />

Pharmaceuticals, Inc; Pfizer Inc; Sanofi-Aventis; and U.S. Surgical.<br />

Some panel members do not accept any support from <strong>in</strong>dustry. The<br />

panel did not regard any potential conflicts of <strong>in</strong>terest as sufficient<br />

reason to disallow participation <strong>in</strong> panel deliberations by any<br />

member.<br />

MS-9


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

References<br />

1. Jemal A, Siegel R, Ward E, et al. <strong>Cancer</strong> Statistics, 2006.<br />

CA <strong>Cancer</strong> J Cl<strong>in</strong> 2006;56:106-130.<br />

2. Blot WJ, Devesa SS, Kneller RW, et al. Ris<strong>in</strong>g <strong>in</strong>cidence of<br />

adenocarc<strong>in</strong>oma of the esophagus and gastric cardia. JAMA<br />

1991;265:1287-1289.<br />

3. Levi F, La Vecchia C. Adenocarc<strong>in</strong>oma of the esophagus <strong>in</strong><br />

Switzerland (letter). JAMA 1991;265:2960.<br />

4. Powell J, McConkey CC. Increas<strong>in</strong>g <strong>in</strong>cidence of adenocarc<strong>in</strong>oma<br />

of the gastric cardia and adjacent sites. Br J <strong>Cancer</strong> 1991;62:440-443.<br />

5. Reed PI. Chang<strong>in</strong>g pattern of esophageal cancer. Lancet<br />

1991;338:178.<br />

6. Powell J, McConkey CC, Gillison EW, et al. Cont<strong>in</strong>u<strong>in</strong>g ris<strong>in</strong>g trend<br />

<strong>in</strong> oesophageal adenocarc<strong>in</strong>oma. Int J <strong>Cancer</strong> 2002;102:422-427.<br />

7. Crew KD, Neugut AI. Epidemiology of upper gastro<strong>in</strong>test<strong>in</strong>al<br />

malignancies. Sem<strong>in</strong> Oncol 2004;31:450-464.<br />

8. Kubo A, Corley DA. Marked regional variation <strong>in</strong><br />

adenocarc<strong>in</strong>omas of the esophagus and the gastric cardia <strong>in</strong> the<br />

United States. <strong>Cancer</strong> 2002;95:2096-2102.<br />

9. Nomura A. Stomach cancer. In: <strong>Cancer</strong> Epidemiology and<br />

Prevention, 2nd edition. Shottenfeld D, Fraumeni JF, eds. New York:<br />

NY. Oxford University Press. 1996:707-724.<br />

10. Corley DA, Buffler PA. Oesophageal and gastric cardia adenocarc<strong>in</strong>omas:<br />

analysis of regional variation us<strong>in</strong>g the <strong>Cancer</strong> Incidence <strong>in</strong><br />

Five Cont<strong>in</strong>ents database. Int J Epidemiol 2001;30:1415-1425.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

11. Park<strong>in</strong> DM, Muir CS. <strong>Cancer</strong> Incidence <strong>in</strong> Five Cont<strong>in</strong>ents.<br />

Comparability and quality of data. IARC Sci Publ 1992:45-173.<br />

12. Kajitani T, Japanese Research Society for the Study of <strong>Gastric</strong><br />

<strong>Cancer</strong>. The general rules for gastric cancer study <strong>in</strong> surgery and<br />

pathology. Jpn J Surg 1981;11:127-145.<br />

13. Fitzgerald RC, Caldas C. Cl<strong>in</strong>ical implications of E-cadher<strong>in</strong><br />

associated hereditary diffuse gastric cancer. Gut 2004;53:775-778.<br />

14. Huntsman DG, Carneiro F, Lewis FR, et al. Early gastric cancer<br />

<strong>in</strong> young, asymptomatic carriers of germ-l<strong>in</strong>e E-cadher<strong>in</strong> mutations.<br />

N Engl J Med 2001;344:1904-1909.<br />

15. Japanese Research Society for <strong>Gastric</strong> <strong>Cancer</strong>. The General<br />

Rules for the <strong>Gastric</strong> <strong>Cancer</strong> Study <strong>in</strong> Surgery and Pathology, 12th<br />

ed. Tokyo: Kanahara Shuppan, 1993.<br />

16. Roder JD, Bottcher K, Busch R, et al. Classification of regional<br />

lymph node metastasis from gastric carc<strong>in</strong>oma. German <strong>Gastric</strong><br />

<strong>Cancer</strong> Study Group. <strong>Cancer</strong> 1998;82:621-631.<br />

17. Leichman L, Silberman H, Leichman CG, et al. Preoperative<br />

systemic chemotherapy followed by adjuvant postoperative<br />

<strong>in</strong>traperitoneal therapy for gastric cancer: A University of Southern<br />

California pilot program. J Cl<strong>in</strong> Oncol 1992;10:1933-1942.<br />

18. Ajani JA, Mayer R, Ota DM, et al. Preoperative and<br />

postoperative comb<strong>in</strong>ation chemotherapy for potentially resectable<br />

gastric carc<strong>in</strong>oma. J Natl <strong>Cancer</strong> Inst 1993;85:1839-1844.<br />

19. Hermanek P, Wittek<strong>in</strong>d C. Residual tumor (R) classification and<br />

prognosis. Sem<strong>in</strong> Surg Oncol 1994;10:12-20.<br />

20. Weber WA, Ott K. Imag<strong>in</strong>g of esophageal and gastric cancer.<br />

Sem<strong>in</strong> Oncol 2004;31:530-541.<br />

REF-1


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

21. Chau I, Norman AR, Cunn<strong>in</strong>gham D, et al. Multivariate<br />

prognostic factor analysis <strong>in</strong> locally advanced and metastatic<br />

esophago-gastric cancer--pooled analysis from three multicenter,<br />

randomized, controlled trials us<strong>in</strong>g <strong>in</strong>dividual patient data. J Cl<strong>in</strong><br />

Oncol 2004;22:2395-2403.<br />

22. Bozzetti F, Marub<strong>in</strong>i E, Bonfanti G, et al. Subtotal versus total<br />

gastrectomy for gastric cancer: five-year survival rates <strong>in</strong> a<br />

multicenter randomized Italian trial. Italian Gastro<strong>in</strong>test<strong>in</strong>al Tumor<br />

Study Group. Ann Surg 1999;230:170-178.<br />

23. MacDonald JS, Smalley SR, Benedetti J, et al.<br />

Chemoradiotherapy after surgery compared with surgery alone for<br />

adenocarc<strong>in</strong>oma of the stomach or gastroesophageal junction. N<br />

Engl J Med 2001;345:725-730.<br />

24. Bonenkamp JJ, Hermans J, Sasako M, et al. Extended lymphnode<br />

dissection for gastric cancer. Dutch <strong>Gastric</strong> <strong>Cancer</strong> Group. N<br />

Engl J Med 1999;340:908-914.<br />

25. Hartgr<strong>in</strong>k HH, van de Velde CJ, Putter H, et al. Extended lymph<br />

node dissection for gastric cancer: who may benefit? F<strong>in</strong>al results of<br />

the randomized Dutch gastric cancer group trial. J Cl<strong>in</strong> Oncol<br />

2004;22:2069-2077.<br />

26. Cuschieri A, Weeden S, Field<strong>in</strong>g J, et al. Patient survival after<br />

D1 and D2 resections for gastric cancer: long-term results of the<br />

MRC randomized surgical trial. Surgical Co-operative Group. Br J<br />

<strong>Cancer</strong> 1999;79:1522-1530.<br />

27. McCulloch P, Nita ME, Kazi H, et al. Extended versus limited<br />

lymph nodes dissection technique for adenocarc<strong>in</strong>oma of the<br />

stomach. Cochrane Database Syst Rev 2004;(4):CD001964.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

28. Mansfield PF. Lymphadenectomy for gastric cancer. J Cl<strong>in</strong> Oncol<br />

2004;22:2759-2761.<br />

29. Degiuli M, Sasako M, Ponti A, et al. Survival results of a<br />

multicentre phase II study to evaluate D2 gastrectomy for gastric<br />

cancer. Br J <strong>Cancer</strong> 2004;90:1727-1732.<br />

30. Sano T, Sasako M, Yamamoto S, et al. <strong>Gastric</strong> cancer surgery:<br />

morbidity and mortality results from a prospective randomized<br />

controlled trial compar<strong>in</strong>g D2 and extended para-aortic<br />

lymphadenectomy--Japan Cl<strong>in</strong>ical <strong>Oncology</strong> Group study 9501. J<br />

Cl<strong>in</strong> Oncol 2004;22:2767-2773.<br />

31. Jansen EP, Boot H, Verheij M, et al. Optimal locoregional<br />

treatment <strong>in</strong> gastric cancer. J Cl<strong>in</strong> Oncol 2005;23:4509-4517.<br />

32. van de Velde CJ, Peeters KC. The gastric cancer treatment<br />

controversy. J Cl<strong>in</strong> Oncol 2003;21:2234-2236.<br />

33. Kooby DA, Suriaw<strong>in</strong>ata A, Klimstra DS, et al. Biologic predictors of<br />

survival <strong>in</strong> node-negative gastric cancer. Ann Surg 2003;237:828-835.<br />

34. Hyung WJ, Cheong JH, Kim J, et al. Application of m<strong>in</strong>imally <strong>in</strong>vasive<br />

treatment for early gastric cancer. J Surg Oncol 2004;85:181-185.<br />

35. Ono H, Kondo H, Gotoda T, et al. Endoscopic mucosal resection<br />

for treatment of early gastric cancer. Gut 2001;48:225-229.<br />

36. Matsumoto Y, Yanai H, Tokiyama H, et al. Endoscopic<br />

ultrasonography for diagnosis of submucosal <strong>in</strong>vasion <strong>in</strong> early<br />

gastric cancer. J Gastroenterol 2000;35:326-331.<br />

37. Yanai H, Noguchi T, Mizumachi S, et al. A bl<strong>in</strong>d comparison of<br />

the effectiveness of endoscopic ultrasonography and endoscopy <strong>in</strong><br />

stag<strong>in</strong>g early gastric cancer. Gut 1999;44:361-365.<br />

REF-2


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

38. Bonenkamp JJ, van de Velde CJ, Kampschoer GH, et al.<br />

Comparison of factors <strong>in</strong>fluenc<strong>in</strong>g the prognosis of Japanese,<br />

German, and Dutch gastric cancer patients. World J Surg<br />

1993;17:410-414.<br />

39. Wieland C, Hymmen U. Megavoltage therapy for malignant<br />

gastric tumors. Strahlenther Onkol 1970;140:20-26.<br />

40. Moertel C, Childs D, Reitemeier R, et al. Comb<strong>in</strong>ed 5fluorouracil<br />

and supervoltage radiation therapy for locally<br />

unresectable gastro<strong>in</strong>test<strong>in</strong>al cancer. Lancet 1969;2:865-867.<br />

41. The Gastro<strong>in</strong>test<strong>in</strong>al Study Group: The concept of locally<br />

advanced gastric cancer: Effect of treatment on outcome. <strong>Cancer</strong><br />

1990;66:2324-2330.<br />

42. Safran H, Wanebo HJ, Hesketh PJ, et al. Paclitaxel and<br />

concurrent radiation for gastric cancer. Int J Radiat Oncol Biol Phys<br />

2000;46:889-894.<br />

43. Anne PR, Axelrod R, Rosato E, et al. A phase II trial of<br />

preoperative paclitaxel, carboplat<strong>in</strong>, 5-FU and radiation <strong>in</strong> patients<br />

with resectable esophageal or gastric cancer (Ca) (abstract). ASCO<br />

Annual Meet<strong>in</strong>g Proceed<strong>in</strong>gs (post-meet<strong>in</strong>g edition). J Cl<strong>in</strong> Oncol<br />

2004;22:4031.<br />

44. Fuchs C, Fitzgerald T, Mamon H, et al. Postoperative adjuvant<br />

chemoradiation for gastric or gastroesophageal adenocarc<strong>in</strong>oma<br />

us<strong>in</strong>g epirubic<strong>in</strong>, cisplat<strong>in</strong>, and <strong>in</strong>fusional (CI) 5-FU (ECF) before<br />

and after CI 5-FU and radiotherapy (RT): a multicenter study. Proc<br />

ASCO 2003;22:257.<br />

45. Ajani JA, Mansfield PF, Crane CH, et al. Paclitaxel-based<br />

chemoradiotherapy <strong>in</strong> localized gastric carc<strong>in</strong>oma: degree of<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

pathologic response and not cl<strong>in</strong>ical parameters dictated patient<br />

outcome. J Cl<strong>in</strong> Oncol 2005;23:1237-1244.<br />

46. Ajani JA, Mansfield PF, Janjan N, et al. Multi-<strong>in</strong>stitutional trial of<br />

preoperative chemoradiotherapy <strong>in</strong> patients with potentially<br />

resectable gastric carc<strong>in</strong>oma. J Cl<strong>in</strong> Oncol 2004;22:2774-2780.<br />

47. Baeza MR, Giann<strong>in</strong>i O, Rivera R, et al. Adjuvant<br />

radiochemotherapy <strong>in</strong> the treatment of completely resected, locally<br />

advanced gastric cancer. Int J Radiat Oncol Biol Phys 2001;50:645-<br />

650.<br />

48. Moertel CG, Childs DS, O'Fallon JR, et al. Comb<strong>in</strong>ed 5-<br />

Fluorouracil and radiation therapy as a surgical adjuvant for poor<br />

prognosis gastric carc<strong>in</strong>oma. Cl<strong>in</strong> Oncol 1984;2:1249-1254.<br />

49. Dent DM, Werner ID, Novis B, et al. Prospective randomized trial<br />

of comb<strong>in</strong>ed oncological therapy for gastric carc<strong>in</strong>oma. <strong>Cancer</strong><br />

1979;44:385-392.<br />

50. Macdonald JS, Smalley SR, Benedetti J, et al. Postoperative<br />

comb<strong>in</strong>ed radiation and chemotherapy improves disease-free<br />

survival (DFS) and overall survival (OS) <strong>in</strong> resected<br />

adenocarc<strong>in</strong>oma of the stomach and gastro<strong>in</strong>test<strong>in</strong>al junction:<br />

Update of the results of Intergroup Study INT-0116 (SWOG 9008).<br />

Presented at the Am Soc Cl<strong>in</strong> Oncol Gastro<strong>in</strong>test<strong>in</strong>al <strong>Cancer</strong>s<br />

Symposium, San Francisco, CA, January 22-24, 2004 (abstr 6).<br />

51. Smalley SR, Gunderson L, Tepper JE, et al. <strong>Gastric</strong> surgical<br />

adjuvant radiotherapy consensus report - rationale and treatment<br />

implementation. Int J Radiat Oncol Biol Phys 2002;52:283-293.<br />

52. Zhang ZX, Gu XZ, Y<strong>in</strong> WB, et al. Randomized cl<strong>in</strong>ical trial on the<br />

comb<strong>in</strong>ation of preoperative irradiation and surgery <strong>in</strong> the treatment<br />

REF-3


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

of adenocarc<strong>in</strong>oma of the gastric cardia (AGC) - report on 370<br />

patients. Int J Radiat Oncol Biol Phys 1998;42:929-934.<br />

53. Pyrhonen S, Kuitunen T, Nyandoto P, et al. Randomised<br />

comparison of fluorouracil, epidoxorubic<strong>in</strong> and methotrexate<br />

(FEMTX) plus supportive care with supportive care alone <strong>in</strong> patients<br />

with non-resectable gastric cancer. Br J <strong>Cancer</strong> 1995;71:587-591.<br />

54. Murad AM, Santiago FF, Petroianu A, et al. Modified therapy with<br />

5-fluorouracil, doxorubic<strong>in</strong>, and methotrexate <strong>in</strong> advanced gastric<br />

cancer. <strong>Cancer</strong> 1993;72:37-41.<br />

55. Glimelius B, Hoffmann K, Haglund U, et al. Initial or delayed<br />

chemotherapy with best supportive care <strong>in</strong> advanced gastric cancer.<br />

Ann Oncol 1994;5:189-190.<br />

56. Scheithauer W, Komek G, Zeh B, et al. Palliative chemotherapy<br />

versus supportive care <strong>in</strong> patients with metastatic gastric cancer: A<br />

randomized trial (abstract). Proceed<strong>in</strong>gs of the Second International<br />

Conference on Biology, Prevention, and Treatment of GI<br />

Malignancy. Koln, Germany, 1995:68.<br />

57. Kelsen DP, Magill G, Cheng E, et al. Phase II trial of etoposide<br />

(VP-16) <strong>in</strong> the treatment of upper gastro<strong>in</strong>test<strong>in</strong>al malignancies<br />

(abstract). Proc Am Soc Cl<strong>in</strong> Oncol 1982;1:96.<br />

58. Lacave AJ, Izarzugaza I, Anton Aparicio LM, et al. Phase II<br />

cl<strong>in</strong>ical trial of cis-dichlorodiamm<strong>in</strong>eplat<strong>in</strong>um <strong>in</strong> gastric cancer. Am J<br />

Cl<strong>in</strong> Oncol 1983;6:35-38.<br />

59. Macdonald JS, Philip SS, Woolley PV, et al. 5-Fluorouracil,<br />

doxorubic<strong>in</strong> and mitomyc<strong>in</strong> (FAM) comb<strong>in</strong>ation chemotherapy for<br />

advanced gastric cancer. Ann Intern Med 1980;93:533-536.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

60. Cull<strong>in</strong>an SA, Moertel CG, Flem<strong>in</strong>g TR, et al. A comparison of<br />

three chemotherapeutic regimens <strong>in</strong> the treatment of advanced<br />

pancreatic and gastric carc<strong>in</strong>oma: Fluorouracil versus fluorouracil<br />

and doxorubic<strong>in</strong> versus fluorouracil, doxorubic<strong>in</strong>, and mitomyc<strong>in</strong>.<br />

JAMA 1985;253:2061-2067.<br />

61. Wils JA, Kle<strong>in</strong> HO, Wagener DJ, et al. FAMTX (5-FU, Adriamyc<strong>in</strong><br />

and methotrexate): A step ahead <strong>in</strong> the treatment of advanced<br />

gastric cancer: A trial of the European Organization for Research<br />

and Treatment of <strong>Cancer</strong> of the Gastro<strong>in</strong>test<strong>in</strong>al Tract Cooperative<br />

Group. J Cl<strong>in</strong> Oncol 1991;9:827-831.<br />

62. Webb A, Cunn<strong>in</strong>gham D, Scarffe JH, et al. Randomized trial<br />

compar<strong>in</strong>g epirubic<strong>in</strong>, cisplat<strong>in</strong>, and fluorouracil versus fluorouracil,<br />

doxorubic<strong>in</strong>, and methotrexate <strong>in</strong> advanced esophagogastric cancer.<br />

J Cl<strong>in</strong> Oncol 1997;15:261-267.<br />

63. Wilke H, Wils J, Rougier P, et al. Prelim<strong>in</strong>ary analysis of a<br />

randomized phase III trial of FAMTX versus ELF versus cisplat<strong>in</strong>/5-<br />

FU <strong>in</strong> advanced gastric cancer (abstract). Proc Am Soc Cl<strong>in</strong> Oncol<br />

1995;14:206.<br />

64. E<strong>in</strong>zig AI, Lipsitz S, Wiernik PH, et al. Phase II trial of Taxol <strong>in</strong><br />

patients with adenocarc<strong>in</strong>oma of the upper gastro<strong>in</strong>test<strong>in</strong>al tract:<br />

The Eastern Cooperative <strong>Oncology</strong> Group (ECOG) results. Invest<br />

New Drugs 1995;13:223-227.<br />

65. Ohtsu A, Boku N, Tamura F, et al. An early phase II study of a 3hour<br />

<strong>in</strong>fusion of paclitaxel for advanced gastric cancer. Am J Cl<strong>in</strong><br />

Oncol 1998;21:416-419.<br />

66. Ajani JA, Fairweather J, Dumas P, et al. Phase II study of Taxol<br />

<strong>in</strong> patients with gastric carc<strong>in</strong>oma. <strong>Cancer</strong> J Sci Am 1998;4:269-274.<br />

REF-4


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

67. Sulkes A, Smyth J, Sessa C, et al. Docetaxel <strong>in</strong> advanced<br />

gastric cancer: Results of a phase II cl<strong>in</strong>ical trial: EORTC Early<br />

Cl<strong>in</strong>ical Trials Group. Br J <strong>Cancer</strong> 1994;70:380-383.<br />

68. E<strong>in</strong>zig AI, Newberg D, Remick SC, et al. Phase II trial of<br />

docetaxel (Taxotere) <strong>in</strong> patients with adenocarc<strong>in</strong>oma of the upper<br />

gastro<strong>in</strong>test<strong>in</strong>al tract previously untreated with cytotoxic<br />

chemotherapy: The Eastern Cooperative <strong>Oncology</strong> Group (ECOG)<br />

results of protocol E1293. Med Oncol 1996;13:87-93.<br />

69. Taguchi T, Sakata Y, Kanamaru R, et al. Late phase II cl<strong>in</strong>ical<br />

study of RP56976 (docetaxel) <strong>in</strong> patients with advanced/recurrent<br />

gastric cancer: A Japanese Cooperative Study Group trial (group<br />

A). Gan To Kagaku Ryoho 1998;25:1915-1924.<br />

70. Kambe M, Wakui A, Nakao I, et al. A late phase II study of<br />

ir<strong>in</strong>otecan (CPT-11) <strong>in</strong> patients with advanced gastric cancers<br />

(abstract). Proc Am Soc Cl<strong>in</strong> Oncol 1993;12:198.<br />

71. Takiuchi T, Ajani JA. Uracil-tegafur <strong>in</strong> gastric carc<strong>in</strong>oma: A<br />

comprehensive review. J Cl<strong>in</strong> Oncol 1998;16:2877-2885.<br />

72. Ajani JA, Mansfield PM, Dumas P. Oral etoposide for patients<br />

with advanced gastric carc<strong>in</strong>oma. <strong>Cancer</strong> J Sci Am 1999;5:112-<br />

114.<br />

73. Koizumi W, Kurihara M, Nakano S, et al. Phase II study of S-1,<br />

a novel oral derivative of 5-fluorouracil, <strong>in</strong> advanced gastric<br />

cancer. For the S-1 Cooperative <strong>Gastric</strong> <strong>Cancer</strong> Study Group.<br />

<strong>Oncology</strong> 2000;58:191-197.<br />

74. Ohtsu A, Baba H, Sakata Y, et al. Phase II study of S-1, a<br />

novel oral fluoropyrimid<strong>in</strong>e derivative, <strong>in</strong> patients with metastatic<br />

colorectal carc<strong>in</strong>oma. S-1 Cooperative Colorectal Carc<strong>in</strong>oma<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Study Group. Br J <strong>Cancer</strong> 2000;83:141-145.<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

75. Maehara Y. S-1 <strong>in</strong> gastric cancer: a comprehensive review.<br />

<strong>Gastric</strong> <strong>Cancer</strong> 2003;6:2-8.<br />

76. Takahashi I, Kakeji Y, Emi Y, et al. S-1 <strong>in</strong> the treatment of<br />

advanced and recurrent gastric cancer: current state and future<br />

prospects. <strong>Gastric</strong> <strong>Cancer</strong> 2003;6:28-33.<br />

77. Kobayashi O, Murakami H, Yoshida T, et al. Trend for better<br />

survival after the <strong>in</strong>troduction of oral fluoropyrimid<strong>in</strong>e, S-1, <strong>in</strong><br />

patients with recurrent gastric cancer (RGC) (abstract). Annual<br />

Meet<strong>in</strong>g Proceed<strong>in</strong>gs (post-meet<strong>in</strong>g edition). J Cl<strong>in</strong> Oncol<br />

2004;22:4174.<br />

78. Ajani JA, Baker J, Pisters PW, et al. CPT-11 plus cisplat<strong>in</strong> <strong>in</strong><br />

patients with advanced, untreated gastric or gastroesophageal<br />

junction carc<strong>in</strong>oma: results of a phase II study. <strong>Cancer</strong><br />

2002;94:641-646.<br />

79. Boku, N, Ohtsu A, Shimada Y, et al. Phase II study of a<br />

comb<strong>in</strong>ation of ir<strong>in</strong>otecan and cisplat<strong>in</strong> aga<strong>in</strong>st metastatic gastric<br />

cancer. J Cl<strong>in</strong> Oncol 1999;17:319-323.<br />

80. Shirao K, Shimada Y, Kondo H, et al. Phase I-II study of<br />

ir<strong>in</strong>otecan hydrochloride comb<strong>in</strong>ed with cisplat<strong>in</strong> <strong>in</strong> patients with<br />

advanced gastric cancer. J Cl<strong>in</strong> Oncol 1997;15:921-927.<br />

81. DeMario MD, Rata<strong>in</strong> MJ. Oral chemotherapy: Rationale and<br />

future directions. J Cl<strong>in</strong> Oncol 1998;16:2557-2567.<br />

82. Humerickhouse RA, Schilsky RL. Thymidylate synthase<br />

<strong>in</strong>hibitors <strong>in</strong> cl<strong>in</strong>ical development. <strong>Cancer</strong> Ther 1998;1:100-113.<br />

83. Kim YH, Sh<strong>in</strong> SW, Kim BS, et al. Paclitaxel, 5-fluorouracil, and<br />

REF-5


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

cisplat<strong>in</strong> comb<strong>in</strong>ation chemotherapy for the treatment of advanced<br />

gastric carc<strong>in</strong>oma. <strong>Cancer</strong> 1999;85(2):295-301.<br />

84. Roth AD, Maibach R, Mart<strong>in</strong>elli G, et al. Docetaxel (Taxotere)cisplat<strong>in</strong><br />

(TC): An effective drug comb<strong>in</strong>ation <strong>in</strong> gastric carc<strong>in</strong>oma.<br />

Swiss Group for Cl<strong>in</strong>ical <strong>Cancer</strong> Research (SAKK), and the<br />

European Institute of <strong>Oncology</strong> (EIO). Ann Oncol 2000;11:301-306.<br />

85. Ross P, Nicolson M, Cunn<strong>in</strong>gham D, et al. Prospective<br />

randomized trial compar<strong>in</strong>g mitomyc<strong>in</strong>, cisplat<strong>in</strong>, and protracted<br />

venous-<strong>in</strong>fusion fluorouracil (PVI 5-FU) with epirubic<strong>in</strong>, cisplat<strong>in</strong>,<br />

and PVI 5-FU <strong>in</strong> advanced esophagogastric cancer. J Cl<strong>in</strong> Oncol<br />

2002;20:1996-2004.<br />

86. Al-Batran S-E, Atmaca A, Hegewisch-Becker S, et al. Phase II<br />

trial of biweekly <strong>in</strong>fusional fluorouracil, fol<strong>in</strong>ic acid, and oxaliplat<strong>in</strong> <strong>in</strong><br />

patients with advanced gastric cancer. J Cl<strong>in</strong> Oncol 2004;22:658-<br />

663.<br />

87. Tsuji A, Morita S, Shima Y, et al. Phase II study of CDDP + S-1<br />

comb<strong>in</strong>ation chemotherapy for advanced and recurrent gastric<br />

cancer (abstract). ASCO Annual Meet<strong>in</strong>g Proceed<strong>in</strong>gs (post-meet<strong>in</strong>g<br />

edition). J Cl<strong>in</strong> Oncol 2004;22:4148.<br />

88. Louvet C, Andre T, Tigaud JM, et al. Phase II study of oxaliplat<strong>in</strong>,<br />

fluorouracil, and fol<strong>in</strong>ic acid <strong>in</strong> locally advanced or metastatic gastric<br />

cancer patients. J Cl<strong>in</strong> Oncol 2002;20:4543-4548.<br />

89. Moehler M, Haas U, Siebler J, et al. Weekly treatment with<br />

ir<strong>in</strong>otecan, fol<strong>in</strong>ic acid and <strong>in</strong>fusional 5-fluorouracil (ILF) <strong>in</strong> patients<br />

with advanced gastric cancer. Anticancer Drugs 2003;14:645-650.<br />

90. Sumpter K, Harper-Wynne C, Cunn<strong>in</strong>gham D, et al.<br />

Randomized, multicenter phase III study compar<strong>in</strong>g capecitab<strong>in</strong>e<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

with fluorouracil and oxaliplat<strong>in</strong> with cisplat<strong>in</strong> <strong>in</strong> patients with<br />

advanced oesophago-gastric cancer: Interim analysis. Proc Am Soc<br />

Cl<strong>in</strong> Oncol 2003;22:257.<br />

91. Bouche O, Raoul JL, Bonneta<strong>in</strong> F, et al. Randomized multicenter<br />

phase II trial of a biweekly regimen of fluorouracil and leucovor<strong>in</strong><br />

(LV5FU2), LV5FU2 plus cisplat<strong>in</strong>, or LV5FU2 plus ir<strong>in</strong>otecan <strong>in</strong><br />

patients with previously untreated metastatic gastric cancer: A<br />

Federation Francophone de <strong>Cancer</strong>ologie Digestive Group Study--<br />

FFCD 9803. J Cl<strong>in</strong> Oncol 2004;22:4319-4328.<br />

92. Shah MA, Schwartz GK. Treatment of metastatic esophagus and<br />

gastric cancer. Sem<strong>in</strong> Oncol 2004;31:574-587.<br />

93. Tian J, Chen L, Wei B, et al. The value of vesicant 18Ffluorodeoxyglucose<br />

positron emission tomography (18F-FDG PET)<br />

<strong>in</strong> gastric malignancies. Nucl Med Commun 2004;25:825-831.<br />

94. Chen J, Cheong JH, Yun MJ, et al. Improvement <strong>in</strong> preoperative<br />

stag<strong>in</strong>g of gastric adenocarc<strong>in</strong>oma with positron emission<br />

tomography. <strong>Cancer</strong> 2005;103:2383-2390.<br />

95. Ott K, F<strong>in</strong>k U, Becker K, et al. Prediction of response to<br />

preoperative chemotherapy <strong>in</strong> gastric carc<strong>in</strong>oma by metabolic<br />

imag<strong>in</strong>g: results of a prospective trial. J Cl<strong>in</strong> Oncol 2003;21:4604-<br />

4610.<br />

96. Jadvar H, Tatlidil R, Garcia AA, et al. Evaluation of recurrent<br />

gastric malignancy with [F-18]-FDG positron emission tomography.<br />

Cl<strong>in</strong> Radiol 2003;58:215-221.<br />

97. Colasanto JM, Prasad P, Nash MA, et al. Nutritional support of<br />

patients undergo<strong>in</strong>g radiation therapy for head and neck cancer.<br />

<strong>Oncology</strong> 2005;19:371-382.<br />

REF-6


®<br />

<strong>Practice</strong> <strong>Guidel<strong>in</strong>es</strong><br />

NCCN <strong>in</strong> <strong>Oncology</strong> – v.1.2007<br />

Recommended Read<strong>in</strong>g<br />

Greene FL, Page DL, Flem<strong>in</strong>g ID, et al. AJCC <strong>Cancer</strong> Stag<strong>in</strong>g<br />

Manual, 6th ed. New York: Spr<strong>in</strong>ger-Verlag, 2002.<br />

Gunderson LL, Burch PA, Donohue JH. The role of irradiation as a<br />

component of comb<strong>in</strong>ed modality treatment for gastric carc<strong>in</strong>oma. J<br />

Infusional Chemo 1995;5:117-124.<br />

Gunderson LL, Sos<strong>in</strong> H. Adenocarc<strong>in</strong>oma of the stomach. Areas of<br />

failure <strong>in</strong> a reoperation series: Cl<strong>in</strong>icopathologic correlation and<br />

implications for adjuvant therapy. Int J Radiat Oncol Biol Phys<br />

1982;8:1-11.<br />

Hallissey MT, Dunn JA, Ward LC, et al. The second British Stomach<br />

<strong>Cancer</strong> Group trial of adjuvant radiotherapy or chemotherapy <strong>in</strong><br />

resectable gastric cancer: Five-year follow up. Lancet<br />

1994;343:1309-1312.<br />

Horn RC. Carc<strong>in</strong>oma of the stomach: Autopsy f<strong>in</strong>d<strong>in</strong>gs <strong>in</strong> untreated<br />

cases. Gastroenterology 1955;29:515-525.<br />

<strong>Gastric</strong> <strong>Cancer</strong><br />

Manuscript<br />

update <strong>in</strong><br />

progress<br />

Version 1.2007, 03/09/07 © 2007 National Comprehensive <strong>Cancer</strong> Network, Inc. All rights reserved. These guidel<strong>in</strong>es and this illustration may not be reproduced <strong>in</strong> any form without the express written permission of NCCN.<br />

<strong>Guidel<strong>in</strong>es</strong> Index<br />

<strong>Gastric</strong> Table of Contents<br />

Stag<strong>in</strong>g, MS, References<br />

Landry J, Tepper J, Wood W, et al. Analysis of survival and local<br />

control follow<strong>in</strong>g surgery for gastric cancer. Int J Radiat Oncol Biol<br />

Phys 1990;19:1357-1362.<br />

McNeer G, Vanderberg H, Donn FY, et al. A critical evaluation of<br />

subtotal gastrectomy for the cure of cancer of the stomach. Ann<br />

Surg 1957;134:2-7.<br />

Papachristou DN, Fortner JG. Local recurrence of gastric<br />

adenocarc<strong>in</strong>oma after gastrectomy. J Surg Oncol 1981;18:47-53.<br />

Wisbeck WM, Becher EM, Russell AH. Adenocarc<strong>in</strong>oma of the<br />

stomach: Autopsy observations with therapeutic implications for the<br />

radiation oncologist. Radiother Oncol 1986;7:13-18.<br />

REF-7

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!