. ..a. . :0. .....1 ... ..qtr . . . . . .. ... L4...04.......v....1<.. .— . . ....o 4 i... ... 4”. 0.0.30.9“. 0. . . 4. 9 . . . .. I... .. .. 9.... .0... .... .. . . . . .0. "0. . 9. ..1u..00~....0 .440 . ..l ..0...0.. ...0 .. . ._ . .0 . . . v. 0 . . . . . . . 0&9‘000900 . . 004 . 0 . 7 . . . .7..‘.7.7....0.404~0£.. . . . .0310... 0.4. . .0. ..0. 70.0... . . . . . .00 00.90.0. . .0 000:”; O0 0 .00 0 o 40300009 I . .. . ..v .. 30.4.0091. . . 9k “000 0 .9 ...4 0.0.9.. o ...1 u n. .0... . . . 300‘. 004. A . .. . . 4 a. o. . ......P.i..-.’Sno.4.:’. . .. . . . ”4.20“”..qu012...(.3.4.»-.. .. o: 4. 4. ...70 040. 04 7 44 .. . . ...0. ...; ... .0. ..ILJ. .0... 700000. 0 407.0..400900:1.\~0 0.. . .9}: ..0 s... . . .. .0000 Q4, . . .1039...— . .. . 3 8 10.4.20”! 0000.59” . . . . . . ... . ... 0. . . . . .0009: 4.4 « .. ...m . . . . . . .. . .. . . . . . . 740. .49“...- u. . . ..04 .. . 9.4). c a 4.4.9“?! . . . ... .. ..... . 7 .. . . 7. .7.. A... .. ...: a . _ . . 7 7 7 . . . . . ..n......:r?.:.9:...~77.. 7 .... ... ..P:.....:.....H.m .2 . . . 000. 0104.000. 0 ... . . .1 0.\ . . . . . 230.0 0.. 49‘ ...0 .14": 00.. .... . ... . .0310 . 7 “.9011. . .90... ...0 . .M30000004..0 .0 4. . 7 0 0 .00.; 0 .....u . 7. . . . . . ....PJ; . .0... 3.....[. .- . 0.. ... 7 . . . 7 . 7 . . 2 . . . 7 . 71....“ 0.0.”. . . . a loom .. 3.: 3.3.00. 3.000;... . . . . . 7 0.0... 4.904..- . . . .‘no..0..d.. .a.. ..4 44 . .. . . .. . . . .00.}...0. . .. . . 30...... o 0:33... ... 0. [.4u0 . 4 0.: .. 00 3.00.900 n . 7 .7 . . 0. n 0 ...... .0 ..a 0040.00 0.9 . 00 .7 9...: 0.... 00. .l . 7 . .10.... {.30 03K. . . . . . . . . . ~ . . 0.09.9: 4.00... . . . 7 9000.0.- Q . ...930. . 0 90 O D 0:. I .10. 90.0.04. V. 490.. ... 00.090. .0072... 90004 . .u .o...0.u n: . 0 $.1..29u.0~$..f._4 ......0’: .000. .40. .0. .00... .040 #9.. ...“, ...90000 0 . 9. O 09000 . 4w. .Pn9Lm $19.09...." .7.-cw. . 0.3L .99."? . 40.”..44.4. ...u 99. ."u400.0.9:.a7 £.lJMk0¥.?0 404 0...“..4. 99.. I“ . . . 00. u . .900 .... . . . . . . . . .. . . . . . . .«. p. . .70."...uv.z.31. ...!on”...94 4 n ”4084...; um . . . _ . . . . 7 . . . . . .. . . . 3.30.4». . .4. .. ...x..u......uz...m$u .mua.u. ...: ...J? ...-.... . 05.300313... 0 .0. 0. 0.0. 000300. 0 ..0. .... . . . . : .. . . . . .. p . 7 . 44.0.“.00. 11...; 4?...u0r702? ...0...0o.<.1..4 ....”h'fl..7..o .. . ...... 0.80 3'90...‘ 3 .00... 0.... 0.0.2”... .... ..I. .7 _ . . . . 7 . . . . 3.. . .930 . .. ..u 39.0.0. . m... . . . o . . t 4 .. .094. 4.0.4 o. .. 0.0.0... . ....1.o.. . 3.0 . . . 7 . . .. . ... .0. ..u._...1-«..0me... ... . .. . .7 3.7.9. 7. . . . . 0...". 3.... 10.4.. . 451...]. 09.0.00. .40.c0.0..:p0 00o .. .43... .9 . .....003 9.. 9 34.. . . . 7 7 . . .. . .00. 4. 0.. ...9 ... 30.0 . .. . {1.00.900 1.. 3 ... .. 04...... . 0 .. 81. S . ..7 423...!09 3.3.0. . 19.1.5." I . . . . . $.04“ 2.409..“ n 7 ... ... a... 6. . .. ... ..0.. 0...: .0 . .. .030: . ...... .7u.0..l. 3.0.0 3.14.0... . .90. L0... . .0 7 . . . . ....o .2. o .0 I. .33... 907 00.0.9.0..9000 00 . . 090. .0 0.00. .n 7 . . . . . 7.44 I. .lnMSJ‘0 n g 0. 4 ..00‘0073 . u . . .. 7 . . . . 7 . . . . . . . . .0 0...“... . . ... .... o. . 7 .. . . . . .. . . . . . . 02:. 1-.9:‘..0 ....‘4090 .00. . ...-0.. 30. v. .....9030. “......9: 0.0. .99. . . . . . . . 9-. 990.0.‘...'..¢l 9.0.0.0 0 ...0: ..0. (00.0020.- . , . . .93.- ...o. 0 . 304.000.30.99 :0 004.099.... . . . . 9.... .01.... .. .99.... .00 .1... 0.0 ... 9'0. 0.0... . .0190... 000. 0 v .0... 0. . S- . ... 1 9.0... .300. 0: 4 . .... 9 - ...-...... 3.4.7.930. . . .vw 04 .0... . 400.."00-0 < .4“ . ~90..- ‘ . . . ...!!‘o .93.? . o0. . c ..r ; . . . _ . . 7. . . “......03590 «Huron-4... 53.9 a. a 4 ."...... . . . . 7 . . . . 4 . 20.0. . .04 a. 4 09 01.0.. 7 . ....4. 0. ("0993‘ ”c.34- 0.1090 ......00: 00400:. 0. .359... ...40.... ......o...... . . . .. . . s .. ...... .2. ... ...}..9! 85.41““. rag Jaut. ...0 .... .2300. . 4 . . .. . . 7 . 7.04.... 2100...}; [9.0.1.8. 404040409..Q.. . . .."ruwu fin.h.000.94§v9yr.vznop.o_m1 . . . . 9001.40 30.. 0...... 4 .. 9,909.2...0.‘ 10..40‘.0..o.,l.404 . . . . . . . 4. .4 .3 . 004 . 06.1934211309 094 2.0... 0. 00.3! .0000 J. .00-000000..0A4 000;: .915 . . . . . . .7 . .. . .... .. ... :7... .0 ..2. .. ...”...t2 3 7...-3.9.3.... 3.4... .32.... .W.....7. 1.». 1 801...}... 0.3.3.9 290.8. 0.0. . . . . u o . u. 2 .20 . 0.... 4 o. .9 .0: 9 0 . 4 0 ... .. .... .3 0 n ....40’? .000.4. 0 ..o. . 7 . . . .. ... .l 0000... u. .0 0.0 . o. .0. .w: . ..«p. . ‘0 . 7 9 0.9th can-“0 09.19.00. . . . 0.. Q”. .0" .0 .504 0:10 .. I» v .....I 91”.... . . .3. ... l ..a .0... ... .0... 9 0,: .0.. .. .... . q . 0 .30....- 00. ......00009. 4 u. a . .0... ..9. 0 0.4.... -0004: .0. 0.0 0'). 09. . .. .‘0: .00.... 0. .09.... O .0 . . . 00 .0. ...«. c. . 0)! .0. I... 02.0”“... '. 941i30‘9100: .0. .00 .0“.-940.01.00.3’49..30....003...‘ . . ...» ...: . . .. 7 . ... . 00. .0 0 0 7. . . . . ,, .00 0. 4 n . 00.0.:- 4.0.’0-... -. 9 .0: . . . . 7. 00?. u: .o 0. .0... ...... 0.00... . .0 0. 0 ... ...8. 1.9.0.9.. 5. -..... .....010 4. .. .. . . 9.9 0009.000 4:0: .0 14.00 0.0.9 0 .0 . 040nv 0.09.0900: .000 00. 0.... 90.1.9... . .970 19.0.: a :9. 00.0.. .... . . . . . . ...ONZ £00 ...... .103... . . . 3. a .74. . . . . .. .. . . . . . .. . . . 7. . "9 .060...“ 0.. 9“ 3900...! 044.909.. 0 ..0.00010. v 00:00 0.. . . .... 7 . . .. . . . . . 7 .. .9900. 0.09. 400.094 . . . . . . 7 . . . . ’. 05.300900390000‘0 0.04. 0 7.... .. .. .. . . . . .. .3. 4.. . 9.0:. .o . ...... o 00 .Oh . .... ..0. . . . . . . . . . . . .. . . . .0 d” 7 .00. .... . . . 9.1.04.1 .0..I . .. . I . .7.! .900... 0. .0... ......0 ‘00. 0 . 0.. 009100 ...-Ivan: . 0. .u 0.000 4 . 1’4 00.. .19...” ... . .0:. 090.0107 .9 . . . . . o. .9. .. .0.. 98 01.909 0.0“ .0033}. .9 90.04 . .. 0 . . .. . ,I09.0=.0o0... . . ...: .«9 .. 79.4.34. .... .0“... 0 7... .........0 . . . _ 7 0... J .0900: .9. 00.0 . . . . . . .0..090o 0.7.39.30 . 4. . ."n..§. . do; .u' 0: .000. 0 00’ “ 00.00.0040.- . . . 7 . . .o 1000.109... 100......0. 0. . . .000 .. 0 90.0 ...}.7 ...-000 :00 0 .00 .9 ......- ..:o... . .....4...7..o......:. 7.. 0000900099000" ......a.0....0 t.“ ‘5)..." . . 9.... ...0 . . . .9. 9. .0 . . . . . . 0...; .0. .9..0P040.. ; . . . . 0. .0 0. 0. .0 . .4... 4.0 0000. . 0 .07 .0. . . 7 . . 0.. 0. 0.0001 '0. 0-. ......lw‘ 200'. o. _ . . .....n9 . 3.. ..90093102 . . . . . . .o...0. .7 . .. . 70...... o. u u . 4. .... .000. 9... I. . . .. . . . ..90.. .0... ..0...0 0 9.00.13.0" . 00.... C 0.4 7 .. . .. . . .. . .. .. . . .. .. . . . . . . 9w. ...... .... . . . . . . I . . . .05.. I 0009... . 9. 0:0 o.70.0...1.0..’l90v0...0?.. . . . 7 . . . . . . . .‘.0.090.0.H0..”. 000’ 7 . . . .. . .. .. .0... o O... .90.. . .. .004. .9 . O; ..1. .. W '9'!" 7' . . .‘ ... .00. .0.’...J¢u. 904)...70\..000000 . . . . . . . _ . . . 0...... . 27.30... ... ...-11.9.9.1.43fi L.u...~‘ . . . . . . . . . . . . . . 0.27.3110: . 7. .00.? . . . . . . . 7 . . _ . _ . . . 30.0. 3.1129! . 109.0 ....091 . . . . . . . . . . . . . . 0.. O 0, . .. . 0.0.0,...00 . .. . ' 7 0 ‘ 9......H. O. 7 . . . .... ... 9.. 0. . 0 0 30.0.. 1.3. .00 . I .. . . . 0.0. w.~....09..>0 .41, 0.90.. . . . . . . 0.0 0:900:13. .. 000 .. . . . . 0.0. 0 2.00.39 ._ .....0... 00.0.... 0. . . 7 7 7 . . .. (”3.02. .0...k1.0.. 7 . . . . . . . v.7; £90054... . 7 7 2 . ..07 70. 9 .300 30.9 .0: ..0 .. o ..0... ...0.. .... ... ‘ . OI .S\.’0:090.0. O 00. ’09.“. .40... 1... .0... 1m:- ‘0. 01.9.. . 0... .0... o. .9...- .....0...0.00 o. .41. 0.. 0”“... 30.0.... 9‘00. .9... .... .9: 2r- ...0 50.0. . .0... .004...s..0u... . .. 00.. 00.000r0 W.-. .. . 7 .9.000I..000‘0.07. . 7 . . . . . . . .. 4. 0. 04......0 04....- . . . . . .. . . . ... 1......0. up . . . .. . . . . . . 0. . . . . . . . . . o o . .07.. . 7 . .‘0. . . . choiu... ...4 . ... .019... . .9 0 . 0 . .l.‘ J. nomnnmfl" .l . ...-4... . . . "1170-90009 1" ...—{€341 - . .4- .... ....4 .. . 00- .009‘ .73 " 03010! 7 O O '09“. .. 7.0 . . v on 49 I000. 00. Wuuaol. c é”...wa.0m A. affix-...“.lwm. . o. .00 \— . 4 V‘. 400 0 .0 .Mnmmfl 4. ’4. 3”“ . .3“ . . _ _. . ... .... in... ...... . 5...... .... ...... .......... ...... .....é..mk.§....._§? 7 fix. air... _ 7 ... r . .. 5; a id‘fi‘bwLF.‘ . .3... I... 0.....3" .1993... ....‘a. ...? Janus... .. ...: v. : “finnima. ......Jfirt hflnafinfidxafivuun... .. .7”..0w........ . ......l.0.1.. .... : ~ _. . . o c. 0.“! 9 J» a .51. J. nNsrfw.1".-r: . ..1 7 . “thbfi It. I «I .u. immodmofl 6......4u79..... ..u... 329.... .... . . 4 9...... ... 31...... ..7 . ..0 ul.' . 0| ..u0119. . v '0 _ 9.1-. V0 009. I olo. 09010.. 0' '00. .0 . 7.. . .40 . .. ...-004 4 “1.... 000.900 404 . .. 0- ..9. v .0 0 00... . .. 0.; 0 u I . . 7. { 0| ’. r! mm LIBRARY 3 Michigan State 7 UNI University This is to certify that the thesis entitled QUANTITATIVE ANALYSIS OF K-RAS MUTATION IN URINE AS AN INDICATOR OF DISEASE STATUS IN PATIENTS WITH STAGE II OR HIGHER COLORECTAL CANCER presented by Shital Darshan Parikh has been accepted towards fulfillment of the requirements for the MS. degree in Clinical Laboratory Sciences 9N, MaonvF‘rofessor’s Signature 1 1/26/08 Date MSU is an Aflinnative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 KrlProj/Acc8PresICIRC/DateDue.indd QUANTITATIVE ANALYSIS OF K-RAS MUTATION IN URINE AS AN INDICATOR OF DISEASE STATUS IN PATIENTS WITH STAGE II OR HIGHER COLORECTAL CANCER BY Shital Darshan Parikh A.THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Clinical Laboratory Sciences 2008 ABSTRACT QUANTITATIVE ANALYSIS OF K-RAS MUTATION IN URINE AS AN INDICATOR OF DISEASE STATUS IN PATIENTS WITH STAGE II OR HIGHER COLORECTAL CANCER BY Shital Darshan Parikh Colorectal cancer (CRC) is the 3rd most commonly diagnosed cancer and the 4th most frequent cause of cancer deaths worldwide. Sequential mutation in various genes can lead to CRC. K-ras mutation is seen in about 50% of CRC patients and is acquired early and remains throughout the process of tumorigenesis. Therefore, detection of mutant K-ras in combination with various screening and surveillance tests may provide early diagnosis, which may enhance the survival rate as well as provide a new tool for determination of prognosis and identification of proper treatment in patients with mutated K—ras. The objective of this study was to develop a validated method to detect and quantitate mutant K-ras in biological specimen. A Restriction Enriched Polymerase Chain Reaction was developed to selectively amplify mutant K-ras, which was then qualitatively detected using gel electrophoresis and quantified using capillary electrophoresis (CE) method. CE method was developed and validated to selectively quantify mutant K-ras at a level as low as 0.05%. ACKNOWLEDGEMENTS I wish to acknowledge my advisor, Dr. John A. Gerlach, for tflxs excellent guidance, supervision, inspiration, and patience throughout the course of my research and thesis write-up. I would like to thank Dr. Gerlach and Michigan State University for providing me research assistantship. I thank all my committee members, Dr. David Thorne, Dr. Kenneth Schwartz, and Dr. Barbara Conley for their helpful suggestions and support during my research. I thank Dr. David Thorne for his valuable suggestions during my course work. I thank Dr. Samuil Umansky for helping with urine isolation method. I thank Dr. Mengiun Wang for providing helpful technical information in isolating DNA from urine specimen. I thank Ann, Ari, Sue, and Michelle for their support. I acknowledge my friends for their cooperation and support. II thank nu! husband, Darshan, for raj; support, love, encouragement, anui immense patience. II thank nu! parents, in-laws, and all my family members for their continuous support throughout nu! study. Also, last 1mm: not least I thank my son, Anish, for giving inspiration and support in thesis write-up. iii TABLE OF CONTENTS LIST OF TABLES ........................................ vii LIST OF FIGURES ....................................... viii RATIONALE ............................................. 1 INTRODUCTION .......................................... 4 Cancer .............................................. 5 Molecular Genetics of Cancer ........................ 6 Gatekeepers .................................... 8 Oncogenes ................................. 8 Tumor Suppressor Gene ..................... 10 Caretakers ..................................... ll Landscaper ..................................... 13 Colorectal Cancer ................................... 14 Epidemiology ................................... 14 Risk Factors ................................... 14 Clinical Background ............................ 17 Sporadic Colorectal Cancer ................ 17 Colorectal Cancer in Inflammatory Bowel Disease ................................... 18 Hereditary Colorectal Cancer .............. 18 Pathogenesis of Colorectal Cancer .............. 20 Screening and Prevention ....................... 23 Staging of CRC ............................. .... 24 TNM (Tumor, Node, Metastasis) Classification... 24 Treatment and Surveillance ..................... 26 RAS Family .......................................... 28 Functional Roles of RAS Proteins in Cancer .......... 3O K-ras Gene and CRC .................................. 31 Presence and Detection of DNA from Serum/Plasma ..... 33 Detection of DNA from Urine ......................... 34 OBJECTIVES ............................................ 36 METHODS ............................................... 37 Preparation of Samples .............................. 38 Positive Control ............................... 38 Negative Control ............................... 38 Serial Dilution of Positive and Negative Controls to Identify Sensitivity of the Method to Detect K-ras ................................ 39 Urine Sample ................................... 39 iv Urine Collection .......................... Ability of Urine Isolation Method to Detect Low Molecular Weight Mutant K-ras sequences ........ DNA Isolation of Samples ............................ Positive and Negative Controls ................. Urine Sample ................................... Primer Preparation .................................. Restriction—Enriched Polymerase Chain Reaction (RE- PCR) ................................................ First-stage PCR ................................ First-round of BstNI Digestion ................. Second-stage PCR ............................... Second-round of BstNI Digestion ................ Polyacrylamide Gel Electrophoresis (PAGE) ........... Detection of Bands of DNA Fragments (Qualitative Method) ............................................. Quantitation of Mutant K—ras Sequences using Capillary Electrophoresis ........................... Matrix File Preparation ........................ Preparation and Capillary Electrophoresis of K- ras DNA Fragment Samples ....................... Data Analysis .................................. Quantitation of Mutant K-ras Sequences in Serial Dilution Assay ..... . ................................ Intra-sample Variability ....................... Inter-sample Variability ....................... RESULTS ............................................... DNA Isolation from Positive and Negative Controls... Detection of K-ras Sequences after RE-PCR and Gel Electrophoresis (Qualitative Method) ................ First-stage PCR and first-round of BstNI Digestion ...................................... Second-stage PCR and second-round of BstNI Digestion ...................................... Ability of Urine Isolation Method to Detect Low Molecular Weight Mutant K-ras Sequences ............. Serial Dilution Assay ............................... Qualitative Analysis of Mutant K-ras Sequences. Quantivative Analysis of Mutant K-ras Sequences in Serial Dilution Assay ....................... Intra-sample Variability .................. Inter-sample Variability .................. DISCUSSION ............................................ DNA Isolation from Positive and Negative Controls... 39 4O 41 41 42 45 46 46 48 48 49 49 5O 51 51 52 53 54 54 55 56 57 57 58 59 6O 62 62 64 67 69 72 73 Detection of K-ras Sequences after RE—PCR and Gel Electrophoresis (Qualitative Method) ................ First-stage PCR and First-round of BstNI Digestion ...................................... Second-stage PCR and Second-round of BstNI Digestion ...................................... Ability of Urine Isolation Method to Detect Low Molecular Weight Mutant K-ras Sequences ............. Serial Dilution Assay ............................... Qualitative Analysis of Mutant K—ras Sequences. Quantitative Analysis of Mutant K-ras Sequences in Serial Dilution Assay ....................... Quantitative Analysis ..................... Intra-sample Variability .................. Inter—sample Variability .................. CONSLUSIONS ........................................... APPENDIX .............................................. Materials ........................................... Instruments ......................................... Figure. 11 Mutant and Wild-type K-ras Sequences during Restriction-Enriched Polymerase Chain Reaction ........................... REFERENCES ............................................ vi 73 73 74 76 77 77 77 77 78 79 80 84 85 89 91 96 Table Table Table Table Table Table Table Table Table Table 10. LIST OF TABLES Risk factors associated with colorectal cancer ....................................... TNM classification of CRC .................... American Joint Committee on Cancer- Union Internationale Contre le Cancer Tumor, Node, Metastasis Staging of CRC .................... PCR primers .................................. Analysis parameters .......................... DNA concentration in SW480 cell-line and PBLs Intra-sample variation ....................... Intra-sample variation. Calcuation of log% mutant and log mean area of data shown in Table 7 ...................................... Inter-sample variation ....................... Inter-sample variation. Calculation of log of % mutant and log mean area on data showed in Table 9 ........................... vfi 16 25 26 46 54 57 68 68 7O 70 Figure Figure Figure Figure Figure Figure Figure Figure Figure Figure Figure. LIST OF FIGURES 1. Formula for the calculation of DNA concentration ............................... 2. A gel photograph after first-stage PCR and first-round of BstNI digestion .............. 3. Photograph of gel electrophoresis after second-stage PCR and second—round of BstNI digestion ................................... 4. The photograph of gel electrophoresis after RE-PCR ...................................... 5. A gel photograph of serial dilution assay after RE-PCR and 12% PAGE ................... 6. A fragment analysis picture of 100%, 75%, 50%, 25%, and 10% mutant K-ras sequences.... 7. A fragment analysis picture of 5%, 1%, 0.5%, 0.25%, and 0.1% mutant K-ras sequences ...... 8. A fragment analysis picture of 0.05%, 0.02%, 0.01%, and 0.002% mutant K-ras sequences.... 9. A plot of log % mutant vs. log mean area (Intra-sample variation) .................... 10. A plot of log % mutant vs. log mean area (Inter—sample variation) ................... 11 Mutant and Wild-type K-ras Sequences during Restriction-Enriched Polymerase Chain Reaction ................................... vfii 44 58 59 61 63 65 66 67 69 71 91 RATIONALE Cancer is aa genetic disease characterized kn! genomic instability. It has been estimated that cells have to acquire at least five to seven successive mutations to allow tumor growth, invasion, and metastasis (Fearon 1990; Boland 1999; Luebeck 2002). Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the fourth most frequent cause of cancer deaths worldwide. In CRC, various cancer susceptibility genes such as oncogenes, tumor suppressor genes and others are involved in tumor initiation and progression in a defined series of stages from normal mucosa to carcinoma (Fearon 1990; Vogelstein 1993). A large number of oncogenes like K-ras (v-Ki-raSZ Kirsten rat sarcoma viral oncogene homolog), Myc [v-myc myelocytomatosis 'viral. oncogene: homolog (avian)], SRC (v- src sarcoma (Schmidt—Ruppin A-2) viral oncogene homolog (avian)], beta—catenin, BRAF (v-raf' murine sarcoma ‘viral oncogene homolog B1) have been identified to play a role in CRC, among all these K—ras is foumd to be most frequently mutated. In CRC, K-ras oncogene has been found mutated in 10-15% of screened adenomas <1 cm, in 30-60% of adenomas >1 cm, and in approximately 50% of adenocarcinomas (Bos J.L. 1989; Kressner U 1998; Fearon E.R. 1990; Boguski M.S. 1993). Current practice uses fecal occult blood test (FOBT), flexible sigmoidOSCOpy, double-contrast barium enema, and colonoscopy (Kahi C.J. 2004). FOBT is a non- invasive test which detects the presence of hidden blood in the stool. Such blood. may come from. anywhere along the digestive tract. Hidden blood in stool is often the first, and in many cases the only warning sign that a person has colorectal disease, including CRC. ffiua main advantages of this test are non-invasiveness and low cost. But the disadvantages include detection of blood in stool, but not its cause, false positive results are also common which may cause anxiety about cancer and lead to unnecessary further tests, and false negative results are also common and may miss disease in its early stages. Due to the non specificity in detecting CRC and chances of false positive and false negative results, it would ibe (n5 interest to develop a test that is more specific and can be used with other techniques to identify CRC more reliably and in early stages. We therefore propose to detect K-ras mutation for screening using urine specimen. Urine is a sample that may be more reliably and safely obtained (in non-invasive manner) and in virtually unlimited amount. Also, molecular testing would be simpler as urine contains less amount of protein for DNA isolation. INTRODUCTION Cancer One of the major characteristics of all higher eukaryotes is the defined lifespan of the organism. Normal cells appear to be mortal, with highly regulated growth and division, and have potential to become terminally differentiated (Trosko 1998; Hanahan 2000). Once the cells are terminally differentiated they can no longer proliferate. Unlike normal cells, cancer cells do not have growth control enui they proliferate indefinitely if provided with adequate nutrients, which suggests that cancer cells may become immortal. Therefore, a cancer can be defined as an unregulated proliferation of cells. Cancer‘ has Gatekeepers Gatekeepers regulate cell proliferation through oncogenes and tumor suppressor genes (Kinzler 1997; Michor 2004). The mutation in both oncogenes and tumor suppressor genes drive the neoplastic process by increasing tumor cell number through the stimulation of cell birth or the inhibition of cell death or cell cycle arrest (Hanahan 2000; Vogelstein 2004). . Oncogenes The oncogenes hypothesis of cancer was proposed based on the discovery that some endogenous viruses contained transforming elements, and the activation of these endogenous transforming elements could cause cancer (Huebner 1969). An oncogene is the mutant form of the proto-oncogene — a cellular gene involved in the control of cell growth and division. These proto-oncogenes can be classified into five broad classes based on their functional and biochemical properties: (a) secreted growth factors [e.g. SIS (simian sarcoma)], (b) cell surface receptors [e.g. ErbB (avian emythroblastosis virus)], (c) components of intracellular signal transduction systems [e.g. the RAS (rat sarcoma) family], (d) DNA-binding nuclear regulatory proteins, including transcription factors [e.g. Myc, JUN (jun oncogene )], (e) components of the network of cyclins, cyclin-dependent kinases and kinases inhibitors that regulate process through the cell cycle [e.g. MDMZ (Mdm2, transformed 3T3 cell double minute 2, p53 binding protein (mouse))]. Oncogenes are frequently activated by gain of functional mutations or fusion with other genes or they are aberrantly expressed due to amplification, increased promoter activity, or protein stabilization (Munger 2002), and hence they play important roles in diverse signaling pathways that are involved in various stages of human cancers-tumors initiation, progression, angiogenesis and metastasis (Michor 2004). An activating somatic mutation in one allele is generally enough to confer a selective growth advantage on the cell. A large number of cellular oncogenes have been identified to play a role in colorectal cancer, such as K-ras, Myc, SRC, beta-catenin, BRAF, and others. It has been reported that approximately 50% of colorectal carcinomas have K-ras gene mutated (Bos (LIN 1989; Boguski M.S. 1993). The RAS family and association of K-ras with colorectal cancer are discussed in details in following sections. . Tumor Suppressor Genes Tumor suppressor genes function to suppress proliferation, promote apoptosis, and maintain integrity of the genome. Mutations in these genes impair this growth- suppressor mechanism resulting in uncontrolled growth. Examples are time genes - Rbl (Retinoblastoma 1J,.jp53, and p16 (cyclin dependent kinase inhibitor 2A). The tumor suppressor genes regulate diverse cell activities including cell cycle checkpoint response, detection and repair of DNA damage, protein. ubiquitination. and (degradationq mitogenic signaling, cell specification, differentiation and migration, and tumor angiogenesis (Sherr 2004). The activation of tumor suppressor genes is usually achieved by deletion of one allele via.aa gross chromosomal event such as loss of heterozygosity (LOH) coupled with an intragenic mutation of the other allele. Most of the identified susceptible genes responsible for familial cancer syndromes are tumor suppressor genes. The inactivating somatic mutations in tumor suppressor genes are also found in sporadic counterpart of the familial cancer syndromes. (Gailani 1996; Ponder 2001; Voglestein 2004). For example, germline mutation of APC predisposes to familial adenomatous polyposis (FAP), Inn: mutation in INK: is also detected in more than 80% of sporadic colorectal cancers. IO The tumor suppressor genes besides APC that are found involved ZUI pathogenesis (Hf colorectal cancer, including p53, DPC1/Smad4 (Deleted in pancreatic carcinoma at loci 1), STKll (Serine/threonine kinase JJJ, PTEN (phosphatase and tensin homolog), DCC (deleted in colorectal carcinoma), Smad4, and others. > Caretakers Caretakers, or stability genes, function in maintaining the genomic integrity of the cell and regulate DNA repair mechanisms, chromosome segregation, and cell cycle checkpoints (Kinzler 1997). Defects in a caretaker gene does not promote tumor initiation, but leads to genetic instabilities that cmmtribute tx> the accumulation of mutations in other genes, including gatekeeper genes that directly affect cell proliferation and survival, thus indirectly promoting tumorigenesis (Lengauer 1998; Rajagopalan 2003; Iwasa 2005). Since deoxyribonucleic acid (DNA) is vulnerable to many types of damage, DNA repair systems are shown to arise early and are highly conserved, in evoluation process (Hoeijmakers 2001). At least four main, partly overlapping DNA repair pathways operate in mammals: the nucleotide-excision repair (NER), base— excision. repair (BER), and Inismatch. repair‘ (MMR) systems 11 are responsible for repairing subtle mistakes made during normal DNA replication or induced by exposure to mutagens (Hoeijmakers 2001; Vogelstein 2004). The homologous recombination and non-homologous end joining (NHEJ) system is responsible for repairing DNA damage involving large proportions of chromosomes, such as double-strand DNA breaks (David 2000; Hoeijmakers 2001; Vogelstein 2004). Each DNA. repair system .is a: complex: biochemical process that requires participation of many genes, and still remains poorly understood. The role of defective DNA repair systems causing tumorigenesis is shown by the existence of human cancer syndromes with germline nmtations in caretaker genes. For example, inborn defects in NER cause syndromes of xeroderma pigmentosum, Cockayne syndrome, and trichothiodystrophy (TTD) are all characterized. by extreme sun sensitivity. Germline mutations in MMR genes, such as hMLHl (human MutL homolog 1 gene), hMSHZ (human MutS homolog 2 gene), and hMSH6 (mutS homolog 63 U3. coli)), are known to predispose to hereditary' nonpolyposis colorectal cancer (HNPCC) (Bandipalliam P 2004). As with tumor suppressor genes, both alleles of caretaker genes generally need to be inactivated for a pathological effect to result. I2 > Landscaper Landscaper defects do not directly affect cellular growth, Inn: generate 6N1 abnormal stromal environment that contributes to the neoplastic transformation of cells (Michor‘ 2004; Iwasa 2005). It is runv well accepted. that cancer develops with malignant transformatbmu of an epitheliuml occurring ‘within. the context carcinomas (Jacoby 1997; Howe 1998). The dysfunction or deregulation of landscaper genes can disrupt normal tissue homeostasis, reduce Ihost immune surveillance and (defense, induce angiogenesis and inflammation, and promote tumor growth and migration (Muller 2004). 13 Colorectal Cancer Epidemiology Colorectal cancer (CRC), a cancer of colon and rectum, is the third most commonly diagnosed cancer and the fourth most frequent cause of cancer deaths worldwide. The World Health Organization estimates more than 11 million new cases diagnosed yearly, with 7 mdllion deaths. CRC is the second leading cause of cancer-related death in the United States; accounting for approximately 148,610 new cases with nearly 55,170 deaths in 2006 (Cancer statistics, 2006). This disease is inore common. in developed countries than o\° developing countries. The lifetime incidence is 5 in developing countries, 1mm: the incidence and mmmtality are now decreasing (Strward 2003; Russo MW 2004; Jemal A 2004). The overall 5-year survival rate in the Unites States exceeds 60%, but is less than 40% in less developed countries (Strward 2003). This variability in disease outcome is proportional to access to specialists and availability of modern drug therapy (Strward 2003). Risk Factors The strongest risk factor for developing CRC is advancing age. The current statistics on CRC indicate that incidence rates rise from 13.2 per 100,000 at age 40-44 to 338.7 per 100,000 at age 75—80 (Ref. SEER data 1975—2001). The overall lifetime risk of CRC is 1.:h1 17 for men and 1 in 18 for women (Cancer Statistics, 2005). Inflammatory bowel disease, a personal or family history of CRC or colorectal polyps, and certain hereditary syndromes are also associated. with, high risk: of developing CRC. Other factors that also contribute to the risk for CRC are: lack of regular physical activity, low fruit and vegetable intake, a low-fiber and high fat diet, obesity, high alcohol consumption and use of tobacco. From a public health perspective, prevention and screening for early diagnosis of CRC is essential to help reduce deaths from CRC. In addition, patients with one diagnosed colon cancer are at risk to develop further colon cancers. l5 Table 1. Risk factors associated with colorectal cancer. Sporadic Colorectal cancer Older age Cholecystectomy (Surgical removal of the gallbladder) Ureterocolic anastomosis Hormonal factors C) Nulliparity (never having carried a pregnancy), 0 late age at first pregnancy, C) Early menopause Environmental factors 0 Diet rich in meat and fat, and poor in fiber, folate, and calcium Sedentary lifestyle Obesity Diabetes mellitus Smoking Previous irradiation Occupational hazards (e.g. asbestos exposure) High alcohol intake OOOOOOO Personal history of sporadic tumors C) History of colorectal polyps c> History of colorectal cancer (risk is l.5-3% for second such cancer in first 5 years) <3 History of small bowel, endometrial, breast, or ovarian cancer Familial colorectal cancer First or second degree relatives with this cancer, criteria for hereditary colorectal cancer not fulfilled: 0 One affected first—degree relative increases risk by two to three fold <3 Two or more affected first-degree relatives increase risk by four to twenty five fold 0 Index case less than 45 years increase risk by three to nine fold c3 Familial history of colorectal adenoma increase risk by two fold I6 Table 1 (cont’d) Colorectal cancer in inflammatory bowel disease 0 Ulcerative colitis o Crohn's colitis Hereditary colorectal cancers 0 Polyposis-syndromes familial adenomatous polyposis (FAP) Gardner’s syndrome Turcot’s syndrome Attenulated adenomatous polyposis coli Flat adenoma syndrome 00000 0 Hereditary non-polyposis colorectal cancer (HNPCC) o Hamartomatous polyposis syndromes c> Peutz-jeghers syndrome 0 JUvenile polyposis syndrome c> Cowden syndrome Source: Strward B.W. 2003 Clinical Background Sporadic Colorectal Cancer Although most cases of CRC are sporadic; genetic and environmental factors also play an important role (Table l) (Strward B.W. 2003). The characteristic of sporadic CRC is the step-wise progression from normal colonic epithelium to malignant growth associated with sequential molecular abnormalities in each step (Kinzler 1996). Fearon et al. showed that colorectal tumors arise as a consequence of the 17 accumulation of activated oncogenes and inactivated tumor- suppressor genes (Fearon E.R. 1990). About 20% of all patients with this cancer are estimated to have some component. of familial risk: without fulfilling‘ the strict criteria for hereditary colorectal cancer (Lynch H.T. 2003). Therefore, family history should always be taken into account when assessing a patient. Colorectal Cancer in Inflammatory Bowel Disease Colorectal cancer accounts for about a third of deaths related to ulcerative colitis, and risk depends on disease duration (2% of affected people by 10 years, 8% by 20 years, and 18% by 30 years), extent of inflammation, presence of primary sclerosing cholangitis, and. backwash ileitis (Itzkowitz S.H. 2004; Krok K.L. 2004). Crohn's colitis is also associated. with increased risk of colorectal cancer; the relative risk is similar to that for ulcerative colitis (Itzkowitz S.H. 2004). Hereditary Colorectal Cancer Roughly 5-10% of all CRCs develop in the setting of defined hereditary cancer syndromes. Hereditary nonpolyposis colorectal cancer (HNPCC, also called Lynch Syndrome) and familial adenomatous polyposis (FAP) are the 18 two :main forms (Lynch H.T. 2003). Various hamartomatous polyposis syndromes are also associated with an jrmmeased risk of such cancer, such as Peutz-jeghers syndrome, juvenile polyposis syndrome, Cowden syndrome (Lynch H.T. 2003; Half E.E. 2004). HNPCC is an autosomal dominantly inherited disorder caused by germline mutations of mismatch repair (MMR) genes. Tumors with HNPCC typically have a molecular characteristic called microsatellite instability. The microsatellite instability is defined as frequent mutations in microsatellites, which are short repeated DNA sequences (Grady W.M. 2003). The penetrance of colorectal cancer in HNPCC is 70-85%. Risk is also increased for tumors of the genitourinary system, stomach, biliary system, pancreas, small intestine, and CNS (Lynch H.T. 2003; Half E.E. 2004; Vasen H.F. 1999). On average, affected. patients develop colorectal cancer by age 44, tumors tend to be right-sided, and, have classical histological features (Bandipalliam P 2004). FAP is an autosomal-dominant disease. In about 80% of affected individuals, a germline mutation can be identified in the adenomatous polyposis coli (APC) gene (Lynch H.T. 2003; Half E.E. 2004). A subset of people with FAP and attenuated FAP, a mild form of FAP which is characterized I9 by the occurrence of fewer colonic adenomas, has biallelic mutations of the rmn: Y’ homolog (MYH) gene (Sieber O.M. 2003; Venesio T 2004). Patients with attenuated FAP typically have mutations at the 5' (proximal to codon 1517) or the 3' end (distal to codon 1900) of the APC gene (Grady M 2003). FAP patients can develop more than 100 colorectal adenomas (50% of patients by age 15 years, 95% by age 35 years); if left untreated, colorectal cancer arises in almost all patients by age 40. Pathogenesis of Colorectal Cancer CRC develops in a series of genetic steps, corresponding with histological progression from normal colonic epithelium to adenomatous dysplasia through microinvasion, adenocarcinoma and, finally metastasis. Two major pathways are known to be involved in the tumorigenesis of CRC. The classical pathway (sometimes called chromosome instability syndrome), is responsible for about 85% of sporadic CRC and is the mechanism of carcinogenesis in patients with FAP, in which cancer cells are characterized by chromosomal instability with mutation of multiple cmcogenes, including KFras, (others oncogenes such as c-myc, c-neu, c-erb-2, c-src are also involved) and many tumor suppressor genes, such as APC, p53, DCC, 20 DPC4/Smad4, and nm32 (Chaung D.C. 2000; Calvert P.M. 2002). Other major pathway that leads to colorectal cancer is via mdcrosatellite instability. Microsatellite instability occurs in approximately 15% of all CRCs and can arise through two mechanisms (De La Chapella A 2003). In the first mechanism, germline mutations of mismatch repair genes (e.g. MLH1, MLH2, and MSH6) in hereditary nonpolyposis colorectal cancer (HNPCC) leads txa deficient DNA—mismatch repair. Nucleotide mismatches occur when two strands of DNA replicate, but almost all such errors are quickly corrected. by a Imolecular proofreading' mechanism. Studies of HNPCC revealed mutation in mismatch repair genes, which encode proteins that repair nucleotide mismatches. Defective mismatch repair presumably facilitates malignant transformation by allowing the rapid accumulation, of Imitations that inactivate: genes ‘with key functions in the cell. The defective MMR genes fail to produce proteins that correct nucleotide mismatches during replication enmi thus promote mutations 1J1 other genes. In addition, genes carrying microsatellite in their coding sequences such as BAX and TGFBR2 are also involved in carcinogenesis via frame shift mutation. 21 In the second mechanism, epigenetic silencing of MLH1 occurs when methylation of the CpG sites in the promoter region of MLH1 silences its transcription and when both the alleles are affected, leads to a mismatch repair deficiency (Herman J.G. 1998). This second mechanism is not heritable and accounts for the majority of all sporadic CRCs that are positive for mfltmosatellite instability (Veigl M.L. 1998; Cunningham J.M. 1998). Additional pathways could exist, for example, the serrated. ‘pathway as vmfld. as distinct. pathways for carcinogenesis of flat and depressed colorectal neoplasms and for carcinogenesis in inflammatory bowel disease (Krok K.I“ 2004; Jass IJJR. 2002; Soetikno IRJ4. 2003; Itzkowitz S.H. 2004). Epigenetic mechanisms such as change in DNA methylation, loss 1 cm, and also, in 50% of the adenocarcinomas (Bos J.L. 1989; Kressner U 1998; Fearon E.R. 1990). The most frequently mutated position in sporadic and familial adenomatous polyposis (FAP) - associated colorectal tumors is codon 12 (B05 J.L. 1989). Because these mutations are acquired early in tumor development (Vogelstein B 1988), and remain through out the process of tumorigenesis, detection of the mutated K-ras in DNA of patients may improve early detection in combination with other screening methods, allowing removal of even small polyps. Testing for mutated K-ras during follow-up after resection from colorectal cancer may provide new tools for detection of recurrence or second primary tumors. Recent evidence implies that tumors with mutated K-ras do not respond to 32 EGFR [epidermal growth factor receptor (erythroblastic leukemia viral (v-erb-b) oncogene homolog, avian)] inhibitors (Van Cutsem 2008; Bokemeyer C 2008; Amado R 2008) and thus may serve to identify appropriate treatments or tn) avoid ineffective treatments, and tflnns may increase survival. Presence and Detection of DNA from Serum/Plasma During apoptosis (programmed cell death), cellular components are dismantled and usually phagocytosed by the macrophages or rmdghboring cells. During the early stages of apoptosis, nuclear [HUI is degraded 1J1 nucleosomes and their oligomers (Wyllie A.H. 1980). Taking into account that in human adults approximately 1011 cells die daily, the amount of released DNA should be approximately 0.6 gram/day (g/day) (Lichtenstein AJV. 2001). However, some chromatin (DNA) degradation products are further degraded into acid- soluble products and are reutilized. within an organism, whereas others escape phagocytosis and appear in the blood- stream. Circulating DNA has been detected in serum and plasma of both healthy and diseased individuals (Anker P 2001). DNA levels III the tflood-stremn presumably reflect the amount (n3 cell death occurring in time whole body and are increased during destructive pathological processes, 33 including cancer and during pregnancy (Chiu R.E. 2004). It has been shown that the serum contains cell-free circulating fragmented DNA iritflue size of nucleosomal DNA (Jahr S 2001). DNA with a cancer “signature” (mutation or hypermethylation) has been found in the plasma or serum of patients with small cell lung cancer (Chen X.Q. 1996), head and neck cancer (Nawroz ii 1996), clear cell renal cancer (Goessl C 1998), pancreatic cancer (Mulcahy H.E. 1998), breast cancer (Chen. X.Q. 1998), hepatocellular carcinoma (Wong IHN 1999), non-small. cell lung cancer (Esteller M 1999), and colorectal carcinoma (Kopreski M 1997). It has also been demonstrated that the mutation found in tumor tissue is the same identical mutations found in plasma or serum of respective patients (Chex X.Q. 1996). The appearance of DNA in plasma from cells dying in a body was also shown by detection of fetus-specific sequences in the plasma of pregnant women (Lo Y.M. 1997; Bischoff F.Z. 1999). Detection of DNA from Urine Experiments on animals demonstrated that DNA fragments of several hundreds base pairs originating from apoptotic cells can cross the renal barrier and preserve their matrix functions. in PCR (Polymerase Chain. Reaction) (Botezatu I 34 2000). This conclushmu was extrapolated IX) humans, as ii; was possible to identify sequences filtered from the blood- stream. through the kidney' barrier into ‘urinary’ DNA“ The possibilities that there are two type of DNA. in ‘urine: excreted DNA, originating from cells dying in various tissues of an organism and second, locally degraded DNA, originating from cells dying 1J1 the urinary tract itself. For example, sequences specific for masculine Y chromosome were detected in urine of women after blood transfusion from male donors and in pregnant women with male fetuses (Botezatu I.V. 2000). Earlier studies demonstrated the possibility of PCR analysis of urinary DNA for diagnosis and monitoring of tumor growth (Su Y.H. 2004). The advantages of detecting mutant sequences in urinary DNA over the analysis of plasma or serum DNA are obvious: it is noninvasive, DNA can be isolated from greater volumes of initial material and is easier to isolate due to low concentration of total proteins (1000-fold lower" protein content). 35 OBJECTIVES The main objective of this research was to develop and validate a method to identify and quantify mutant K-ras in biological specimen. The specific objectives of this research are: > Isolation of DNA . Isolation of low molecular DNA from urine sample 0 Isolation of High molecular DNA from SW480 cell line and Peripheral Blood Leucocytes > Adaptation of qualitative method for the detection of mutant K-ras sequences > Identification of the lowest concentration of mutant K-ras that can be detected qualitatively using RE-PCR and gel electrophoresis by serially diluting mutant K- ras with wild-type K-ras > Develop and validate a method to quantitate mutant K- ras using a Capillary Electrophoresis 0 Design an anti-sense fluorescent primer to quantify mutant K-ras using fluorescence 36 METHODS 37 All the materials and equipment mentioned in this section are listed in the appendix. )‘9 Preparation of Samples . Positive Control Genomic DNA from SW480 cell line (American Type Culture Collection (ATCC)) possessing ea mutation 1J1 codon 12 of the K-ras proto-oncogene was used as a positive control. SW480 cell line cells were obtained in suspension. The suspension was thawed at 37°C in a water—bath (Precision Water Bath) for approximately 5 minutes. The suspension was then centrifuged for 5 minutes at 300 x gravity (g) in a 1.5 mL microcentrifuge tube and the supernatant was removed and discarded. The cell pellet was resuspended in PBS (Phosphate Buffered Saline), pH 7.4 to a final volume of 200 uL. The isolation was carried out using the method described below. 0 Negative Control Genomic DNA from Peripheral Blood Leucocytes (PBLs) was used as a negative control for K-ras codon 12. 38 The whole blood was collected in ACD (Acid Citrate Dextrose), yellow-top tube. The tube was centrifuged at 655 x g tin: 10 minutes, and 200 in; of buffy coat (leucocytes) was used for DNA isolation as described below. 0 Serial Dilution of Positive and Negative Controls to Identify Sensitivity of the method to Detect R-ras The serial dilution assay was performed by diluting SW480 cell line (100% mutant) used as positive control with PBLs (100% wild-type) used as negative control. The dilution range for SW480 cell line was 100, 75, 50, 25, 10, 5, 1, 0.5, 0.25, 0.1, 0.05, 0.02, 0.01, and 0.002%. The RE- PCR was performed (discussed in the sections to follow) on all dilution samples, negative» control (PBLs, 100% wild- type), and blank control of PCR” 11 12% PAGE was performed (discussed in the sections to follow) after RE-PCR. The procedure is performed in triplicate. o Urine Sample 0 urine Collection Twenty five 11) fifty mulliliter nmn of fresh urine was collected (the first void of the day was not used) in a sterile container and immediately' mixed with appropriate 39 volume of 0.5 mole per Liter (M) EDTA (Ethylenediaminetetraacetic acid), pH 8.0 to achieve a final EDTA concentration of 10 millimole per Liter (mM). This concentration of EDTA is known to inhibit nuclease activity (Milde A 1999). Urine samples were stored in 15 mL conical polypropylene screw cap tubes in 10 ml aliquots at -70 °C (degree centigrade). The tubes were thawed and centrifuged (Sorvall TC6 tabletop centrifuge) at 200 x g for 5 minutes and placed on ice before isolation. The urine samples were obtained from a volunteer. Ability of Urine Isolation Method to Detect Low Molecular NeightZMutant K-ras Sequences To evaluate the ability of the method to detect K-ras in urine sample and to determine the sensitivity of the method following samples were prepared: Eight 10 mL aliquots of same urine sample, negative for mutant K-ras were thawed. A different amount (80 uL, 15 uL, 5 uL, 1 uL, 0.1 uL, 0.01 (mg and 0.001 tun of SW480 cell line first round PCR (discussed in the sections to follow) products were added tx> the seven of time eight urine sample tubes. The tubes were mixed and centrifuged at 200 x g for 5 minutes and placed on ice. 40 '> DN3.Isolation of Samples 0 Positive and Negative Controls ® The QIAamp DNA Mini Kit, designed to isolate high molecular weight DNA, was used to obtain genomic DNA from PBLs and SW480. Twenty micro—liter of QIAGEN protease (provided with kit) and 200 in; buffer AL U1 cell lysis buffer, provided with kit) was added to 200 uL of cells or buffy coat. The tube was 'vortexed (Vortexer, Type 16700 Mixture) for 15 seconds, centrifuged, and the mixture then was incubated at 56°C (Precision Water bath-micro) for' 10 minutes. After incubation, the tube was centrifuged, and precipitation of DNA was achieved by adding 200 uL of ethanol and vortexing the tube for 15 seconds. Tube was then centrifuged and the mixture was transferred to the QIAamp spin column (in a 2 mL collection tube, provided with kit) and centrifuged at 9295 x g for 1 minute. The column was placed in a new clean 2 mL collection tube, 500 uL of wash buffer AWl (provided with kit) was added to the column and centrifuged at 9295 x g for 1 minute. The column was placed in a new clean 2 mL collection tube, 500 In; of wash buffer AW2 (provided with kit) was added to the column and centrifuged at 9295 x g 41 for 3 minutes. The column was placed in a clean 1.5 mL UV- treated (90 seconds at 254 nm) (GS Gene Linker UV chamber) microcentrifuge tube and 200 uL of buffer AE (elution buffer, provided. with kit) was added. to the column and allowed to incubate for 5 min at room temperature. The DNA was collected by centrifugation at 9295 x g for 1 minute. The GeneQuant RNA/DNA Calculator was used to measure the optical density at 260nm. Five micro-liter of DNA was diluted to 200 (H; with sterile deionized distilled water. The protein contamination, purity, and 260nm/280nm ratio were also recorded. The cancentratbmn of double stranded, high molecular weight DNA was calculated using equation 1. The DNA samples from SW480 cell line and PBLs were stored at —20°C until further use. 0 ‘Urine Sample Fragmented DNA that appears in urine due to apoptosis has low lmolecular' weight than. the high imolecular‘ weight genomic DNA from the positive and negative controls. Therefore, a different method was used to isolate fragmented DNA in urine than the isolation method used for positive and negative control mentioned in the following sections. DNA was isolated as described previously (Botezatu I 2000; Ying—Hsirl Su 2004) with some lmodifications. The 42 changes to the original method were obtained via personal communication with Samuil Umansky (Xenomics, Inc.). The isolation method is described below. A. 20 mu; of 6 b4 GITC (Guanidine Isothiocyanate) was added t1) 10 mfll of urine i11 a 50 Hui conical polypropylene screw cap tube and mixed vigorously; One mL of resin (Wizard® Pius Minipreps DNA Purification System) was added and the tube was incubated for 4 hours at room temperature with gentle mixing. The resin was pelleted by centrifugation at 200 )(i; for 10 mdnutes. The supernatant was carefully aspirated and discarded. The resin pellet was resuspended in 1 mL of 4 M GITC, and transferred into a 1.5 mL microcentrifuge tube. The tube was centrifuged in a microcentrifuge (Sorvall Biofuge® Pico) at 9295 x g for 1 minute. The supernatant was removed and discarded. The pellet was resuspended in 1 mL of column wash buffer (Wizard® Plus Minipreps DNA Purification System) and suspension was transferred 11) a Promega Wizard mdnicolumn (Wizard® Plus Minipreps DNA Purification System). The DNA was separated from the suspension by applying vacuum. The column was washed with. 2 'mL of column wash buffer. To remove residual washing buffer from the column, the column- tube assembly was centrifuged in a mdcrocentrifuge at 9295 43 x g for 3 minutes. The column was placed in a clean 1.5 mL UV-treated (90 seconds tat 254 :nanometer (nmn) microcentrifuge tube and 100 microliter (uL) of sterile deionized distilled water was added to the column and allowed to incubate for 5 min at room temperature. The DNA was collected by centrifugation at 9295 x g for 1 minute. The GeneQuant RNA/DNA Calculator was used to measure the optical density at 260 nm. Fifty microliter of DNA was diluted to 200 (n; with sterile deionized distilled water. The protein contamination, purity, and 260nm/280nm ratio were also recorded. The concentration of double stranded DNA (dsDNA) was calculated using formula mentioned in Figure 1. The DNA samples were stored at -20°C until further use. Formula for Double stranded DNA DNA ug/uL = [ODum* X 50(ug/mL) X DF** X l mL)]+ 1000 uL *ODmm = Optical density at 260 nm **DF = Dilution factor An optical density at 260nm of 1.0 corresponds to 50ug of DNA per mL for dsDNA. Figure 1. Formula for the Calculation of DNA Concentration. 44 > Primer Preparation Two primer sets were used for the Polymerase Chain Reaction (PCR). The sense and antisense primers of primer set I and sense primer of primer set II were published sequences (Su YH 2004). The antisense primer in primer set 11 was developed specifically for this research project. The antisense primer in the primer set II was labeled with 6-FAM (6-carboxyl fluorescein) at 5’ end. Incorporation of 6-carboxyl fluorescein. imparted. the fluorescence jproperty needed in the detection and quantification of K-ras using the capillary electrophoresis. Capillary electrophoresis was performed using ABI PRISM® 310 Genetic Analyzer. Primers were synthesized. at time Macromolecular Structure Facility at Michigan State University. The primer sequences of both the primer sets are listed in Table 4. The primers were dried under vacuum.ijlaa SpeedVac SC100 system. Dried primers were reconstituted in (Kmarm; of sterile deionized distilled water and stored at -20°C. 45 Table 4. PCR Primers. Primer Set I Sense* GCTCTTCGTGGTGTGGTGTCCATATAAACTTGTGGTAGTTGGACCT Antisense* CKTCTTCGTGGTGTGGTGTCCCGTCCACAAAATGATTCTGA Primer Set II Sense* ACTGAATATAAACTTGTGGTAGTTGGACCT Antisense* is*6'FAMCTGAAGTCCACAAAATGATTCTGAATTAGC *The primer sequence is in 5’ to 3' form. ** The antisense primer 1J1 the primer set IKE was labeled with 6—FAM (6-carboxyl fluorescein) at 5’ end. )> Restriction-Enriched Polymerase Chain Reaction (RE-PCR) K-ras mutation was detected irlea two-stage PCR assay using selective restriction enzyme digestion of an artificially created recognition site t1) distinguiSh wild- type and. mutant K-ras sequences. Mutant K-ras sequences were enriched by RE-PCR. o First-stage PCR PCR amplification was completed in 0.2 mL thin walled reaction tube with a total volume of 50 (AL. A master mix 46 containing all the PCR reagents except for the DNA template was prepared. The reaction mixture includes 1X GeneAmp PCR Buffer, 1.25 Units of AmpliTaq Gold DNA polymerase, 200 uM of each <1f the deoxyadenosine triphosphate (dATP), deoxycytosine triphosphate (dCTP), deoxyguanosine triphosphate (dGTP), deoxythymine triphosphate (dTTP), and 0.9 in; of each of time sense and antisense primers (primer set I, Table 1). For positive and negative controls, DNA templates for SW480 or PBLs at a concentration of 0.1 pg were added. For urine samples the amount of DNA present in 0.2 mL of urine was used as DNA template. Also, a blank was included in each PCR assay, which contained only sterile deionized distilled water instead of DNA template. In each case the final volume of 50 (AL was adjusted with sterile deionized distilled water. Hot start (Perkin Elmer) PCR method was used to produce higher yields of specific PCR amplified products. PCR amplification was performed with the GeneAmp® PCR System 9700 with an initial incubation at 95°C for 15 minutes. Followed by 15 cycles, each cycle included following steps; a) denaturation at 94°C for 48 seconds, b) annealing at 56°C for 90 seconds, and c) extension at 72°C for 2 minutes. A final extension, 72°C for 5 mdnutes, was done to ensure complete amplification of PCR products with 47 a 4°C temperature hold if the PCR product was to remain in the thermal cycler. These first 15 cycles of PCR amplified both wild-type and mutated DNA and introduced an artificial BstNI site to the 5' end of amplified product derived from wild-type K-ras sequences. 0 First-round of BstNI Digestion The restriction endonuclease enzyme, BstNI [sourcez Bacillus stearothermophilus N (D. Comb)], was used to digest artificially created recognition site :hi wild-type K-ras sequences. After the first-stage PCR, the amplified products were digested with BstNI restriction endonuclease enzyme to eliminate amplified products derived from wild-type K-ras sequences. The mixture of 10 uL of first-stage PCR product, 10 Units of BstNI enzyme, and 1X NEBuffer 2 in a total volume of 20 uL (adjusted with sterile deionized distilled water), was incubated at 60°C for 3 hours. 0 Second-stage PCR PCR amplification was completed in 0.2 ml thin walled reaction tube with a total volume of 50 uL. A master mix containing all the PCR reagents except for the DNA template was prepared. The reaction mixture includes 1X GeneAmp PCR 48 Buffer, 1.25 Units of AmpliTaq Gold DNA polymerase, 200 uM of each of dATP, dCTP, dGTP, and dTTP, and Eilfl; of sense and 7.5 uL of antisense primers (primer set II, Table 1). One micro-liter of the first-round of BstNI products was added as DNA template for second-stage PCR. The final volume of 50 uL was adjusted with sterile deionized distilled water. Hot start PCR method was used and PCR amplification was performed with the GeneAmp® PCR System 9700 with an initial incubation at 95°C for 15 munutes. Followed by 35 cycles, each cycle included following steps; a) denaturation at 94°C for 48 seconds, b) annealing at 56°C for 90 seconds, and c) extension at 72°C for 2 munutes. A final extension, 72°C for 5 minutes, was done to ensure complete amplification of PCR products with a 4°C temperature hold if the PCR product was to remain in the thermal cycler. o Second-round of BstNI Digestion 10 uL of second-stage PCR product, 1 X NEBuffer 2, 10 Units of BstNI enzyme, and 16 uL of sterile distilled water were mixed and incubated at 60°C water-bath for 5 hours. 49 S> Polyacrylamide Gel Electrophoresis (PAGE) The final BstNI digestion products were separated by ® electrophoresis using a Mini-PROTEAN II Cell electrophoresis chamber eumi a Model 1000/500 Power Supply. A 12% polyacrylamide gel (19:1 acrylamide to bisacrylamide ratio, 1X TAE) with the dimensions of 75 mm x 85 mm x 0.75 Imn was used. A 10 in; of final BstNI digested product was mixed with 2 in; of 5X nucleic acid sample loading buffer and loaded on the gel along with one lane of molecular weight marker which contained 50-500 bps DNA fragments. The samples were electrophoresed at 150 volts for 1 hour. I> Detection of Bands of DNA Fragments (Qualitative Method) Ethidium bromide dye was used to stain DNA fragments. One drop of ethidium bromide (10 milligram per milliliter (mg/mL)) was diluted in at least 500 mL of water. The gel was placed into diluted ethidium bromide solution and incubated with gentle rotation (American Rotator V) at room temperature for about 10 minutes. The gel was washed thrice with water to remove excess stain on the gel. DNA bands were 'visualized. using' Chromato-Vue Transilluminator Imodel TS-36 at the wavelength of 254 nm. The gels were photographed with type 667 black and white Polarohfa film 50 using a Fotodyne FCR-lO camera. Film was exposed to gel with UV light for one second at f=8 and allowed to develop for one minute. > Quantitation of Mutant R-ras Sequences using Capillary Electrophoresis DNA fragments after the second-round of BstNI enzyme digestion were used for K-ras quantitation. The quantification of mutant K-ras sequences was done by capillary electrophoresis using ABI PRISM® 310 Genetic Analyzer. The combination of the DS-30 dye set and virtual filter set D was used for analysis of all samples. 0 Matrix File Preparation The matrix file contains the information necessary for software to correct the spectral overlap of the dyes in the virtual filter set. The matrix file was generated from a separate matrix standards run, and contains information about how much of the collected light falling on a filter is due to the intended light emission and how much is contaminating light. To generate matrix file, a matrix standard set (DS-30) was used which contains 6-FAM (6-carboxyfluoresein), HEX 51 (4, 7, 2’, 4’, 5', 7’,-Hexachloro-6-carboxyfluorescein), ROX (6-Carboxy-X-rhodamine,), and. NED (Applied. Biosystems Proprietary) fluorescent dyes. Each fluorescent dye emits a continuous spectrum of light upon laser excitation. The matrix standards preparation and the electrophoresis on the ABI PRISM 310 Genetic Analyzer occurred as follows: 2 uL of each of the matrix dyes (6-FAM, HEX, NED, and ROX) was added to 24 uL of deionized formamide in MicroAmp 0.2 mL sample tubes. The mixture was mixed by vortexing and centrifuging for 5 seconds, denatured (DNA Thermal Cycler) at 95°C for 5 minutes and chilled on ice for 5 minutes. The matrix standards were injected ill the capillary by electrokinetic injection for 53 seconds and electrophoresed at 15 kilo Volt (kV) for 24 minutes in Performance Optimized Polymer (POP-4“”jpolymer) with 51 run temperature of 60°C. The matrix standard run data were collected using ® the .ABI PRISM 310 data collection software ‘v3.0.0. The data collection software generated the sample file of each matrix standard. The sample files of each matrix standard were used to create a matrix file using ABI PRISM® GeneScan® Analysis Software v3.7 as described in the user’s manual (Ref. Applied Biosystems). 52 0) Preparation and Capillary Electrophoresis of K-ras DNA Fragment Samples The DNA fragments from the second—round of BstNI digestion were diluted appropriately with deionized distilled water. The appropriate amount of diluted sample ® and 0.5 uL of GeneScannt350 ROX internal lane size standard (vortexed and centrifuged for 5 seconds) were added to 12 (AL of deionized formamide in MicroAmp 0.2 mL sample tube. The mixture was vortexed and centrifuged for 5 seconds, denatured at 95°C for 5 minutes and chilled on ice for 55 minutes. Samples were injected 1J1 a capillary' by electrokinetic injectbmn for 5 seconds and electrophoresed at 15 kilo Volt (kV) for 24 minutes in Performance Optimized Polymer (POP-4TM polymer) with.aa run temperature ® of 60°C. The data were collected using the ABI PRISM 310 data collection software application v3.0.0. The sample file for appropriate sample was created by data collection software. > Data Analysis The electrophoresis results were analyzed ‘using .ABI PRISM® GeneScan® Analysis Software v3.7. The analysis parameters for data processing are described in Table 5. 53 The peaks were interpreted when greater than or equal to 50 relative fluorescence units (RFUs). The matrix file was applied to the samples before analyzing the data” The GS350-250 size standard parameter was used for size calling of DNA fragments. Size calling, peak height, and peak area of samples were determined. Table 5. Analysis Parameters Analysis Parameters values Analysis range 2500 to 4000 Data points Size Call range 35 to 150 base pairs Data Processing :Smooth options None Size Calling method Local Southern Method Peak Detection: (3 Peak Amplitude Thresholds: B 50 Y 50 (3 50 R 50 (3 Minimum Point Half Width 2 Points <3 Polynominal Degree 3 C) Peak Window Size 15 Points c> Slope Threshold for Peak 0.0 Start 0 Slope Threshold for Peak 0.0 end Baselining: BaseLine Window Size 51 Points Quantitation of Mutant K—ras Sequences in Serial Dilution Assay 54 o Intra-sample variability The second BstNI digestion products of RE—PCR from the serial dilution assay (described earlier) were taken to perform the quantitative analysis of mutant K-ras sequences using capillary electrophoresis as described above. The procedure was performed using the same sample three times to determine the intra-sample variability. o Inter-sample variability The second BstNI digestion products of RE-PCR from the serial dilution assay ens described earlier were taken to perform the quantitative analysis of mutant K-ras sequences using capillary electrophoresis as described above. The procedure was performed in triplicate; three different sample sets were prepared. to determine the inter-sample variability. 55 RESULTS 56 DNA Isolation from Positive and.Negative Controls The concentration of DNA isolated from SW480 cell line (positive control), PBLs (negative control), and urine is shown in Table 6. The optical density at 260nm and 280 nm measured using spectrophotometer and 260nmz280nm ratio was calculated. Table 6. DNA Concentration in SN480 cell-line and PBLs on at on at Ratio conc.:::iti° 260 nm 280 nm 260 nut/280 nm . n In nqlnL SW480.Cell‘ 0.059 0.031 1.879 0.118 line PBLs 0.160 0.085 1.892 0.320 Urine 0.038 0.021 1.815 0.0076 Detection of R-ras Sequences after RE-PCR and Gel Electrophoresis (Qualitative Method) The SW480 cell line (positive control), PBLs (negative control), and normal urine negative for mutant K-ras were amplified by RE-PCR as described in materials and 10 uL was electrophoresed on a 12% PAGE as described previously. 57 o First-stage PCR and First- round of BstNI Digestion The results (n? gel electrophoresis following first- stage PCR and first-round of BstNI digestion are shown in Figure 2. Figure 2. A gel photograph after first-stage PCR and first- round of BstNI digestion. Well 1 — molecular ruler (50-500 bps) Wells 2, 4, and 6 — First PCR products of SW480, PBLs, and normal urine negative for mutant K-ras sequences, respectively (125 bps) Wells 3, 5, and 7 — SW480, PBLs, and normal urine negative for mutant K-ras, respectively, after first-stage PCR followed by first round of BstNI digestion Well 8 - negative control for PCR (water, no DNA) The first-stage PCR amplifies both mutant and wild- type K-ras sequences; the amplified fragment size was 125 bps. The BstNI digestion cut the amplified wild-type K-ras sequences in 85 and 40 bps fragments. 58 o Second-stage PCR and Second-round of BstNI Digestion The results of gel electrophoresis following second- stage PCR and second-round of BstNI digestion are shown in Figure 3. Wells: 1 2 3 4 5 6 7 8 <—-30 bps Figure 3. A photograph of gel electrophoresis after second- stage PCR and second-round of BstNI digestion. Well 1 - molecular ruler (50-500 bps) Wells 2, 4, and 6 - second PCR products of SW480, PBLs, and normal urine negative for mutant K-ras sequences, respectively (92 bps) Wells 3, 5, and 7 — K-ras fragments after second BstNI digestion of SW480 (92 bps), PBLs (62 and 30 bps), and normal urine negative for mutant K-ras (62 and 30 bps), respectively Well 8 - negative control for PCR (water, no DNA) The fragment sizes of amplified products after second- stage PCR were 92 bps. The second-round of BstNI digestion cut the amplified wild-type K-ras sequences into 62 and 30 bps fragments. 59 Ability of Urine Isolation Method to Detect Low Molecular weight Mutant K-ras Sequences Gel electrophoresis data for the isolation of 125 bps fragments of SW480 cell line first-stage PCR by urine isolation method and RE-PCR is shown in Figure 4. 6O Amzo 0c noumzv mum you Honucoo obflpmmmc I m Ham: .>Ho>auomamon mucmfimmum man mNH man goes: monsoone mom momomupmuno mans demo omezm no as Hoo.o new .4: Ho.o .q: H.o .q: H .4: m .q: me can om cue: ooxHE moan: HmEuoc Eon“ UoDMHOmH mzo mum me can .NH .HH .OH .> .m .m maamz manlx Denyse now o>apmmoc moans Hmfinoc v Hams meme m cam m maamz mafia Haoo owvzm N Haw: Amen oomuomv Hesse Hassomsos A name .momlnm Hound edeouosmouuUOHo HOD Ho nmiumouonm GAB .v OHSUHH man om.|v won Noile - .r#:» mob mmlv. all I I} l} . MH NH HH OH m m b o m v m N H "maamz 61 Serial Dilution Assay o Qualitative Analysis of Mutant R-ras Sequences The serial dilution assay was performed to determine the sensitivity of RE—PCR method by diluting SW480 cell line (positive control) with PBLs (negative control) to obtain different mutant K-ras concentration as described previously. The gel photograph was obtained after 12% PAGE followed by RE-PCR as showed in Figure 5. 62 moocmsomm NH coooo mmnIx How momuloaaz moofi on oonnomou mqmm moocosomm NH coooo mmuIM How accuse wooa ou commemon omvzm Amzo oc noumzv mom mo Houucoo o>aumooz I m Ammo oomlomv menme Danae: amasooaofi I 232 .noam was can monies uouua manna coauuaao Hanna» mo smiuuouonm Hoe ¢_.m masons man om o n .. .aitl ail.ellpumxllei}_rtis use» 4 m o No . . . . . man Nm IIV.. .. . I... .. 33?. 3:1... 35.5 13.91 } 3 i. I! _ . If... A -15.... fl... 5... a: ... . 232 name omvzm Noo.o Ho.o No.0 mo.o To mN.o m m.o H m 0H mN om me 232 a a fl. l\ {II .I\ .I..._ . ... lJ\I .l/\II pawns: w as .33 z woes Damon: M. 63 0 Quantitative Analysis of Mutant K-ras Sequences in Serial Dilution Assay Quantitative analysis of the mutant K-ras was ® performed using ABI PRISM GeneScan Analysis software. The capillary electrophoresis followed by RE-PCR was performed as described earlier in serial dilution assay. Figures 6-8 show the fragment analysis by ABI PRISM® GeneScan Analysis software \7 3.7. Y-axis indicated fluorescence intensity, while the X-axis indicated base pair size. 64 FBMWWWWB‘ _1 5 U ‘ 7 5 ‘ A 1 0 0 T :3 _,.-'--.E A .-"‘-. :"f‘ '. 1 O O % M ".2 If g. , - 1 ;;L E; 7 5 % M a: 5 O % M - I .__ 1 _ _ 1 :1: 2 5 % M = 1 0 % M MMIIH~174flM II .E‘Culfll Internal size standard (size in nucleotides) I ) Primer peak Wild—type K-ras sequences peak [:1 Mutant K-ras sequences peak Figure 6. A fragment analysis picture of 100%, 75%, 50%, 25%, and 10% mutant K-ras sequences. .All the peaks in different serial dilution samples were aligned by fragment size. 65 INCH-IF,“ ""3"“ I'“ “id-3’1 £4.0mM-h‘e". dot a III-'1‘ M. Mewm.wwuenws “ ‘-"' 'fi "'" “ " ' "‘ :1“ .- . —“ T ' '2' X] Efl-I‘Edl Bond Sarah's” Y- undo- flab A A I 5%M 1%M 0.5% M 0.1% M ‘mlgmoummse— lac-fienn-woj .i a: "38m 5) Internal Size standard (Size in nucleotides) ee F \ J I \ 0 CI Primer peak Wild-type K-ras sequences peak Mutant K-ras sequences peak Figure 7. A. fragment analysis picture of 5%, 1%, 0.5%, 0.25%, and 0.1% mutant K—ras sequences. All the peaks in different serial dilution samples were aligned.by fragment size. 66 anwmmw—mu Ally uni 50 75 100 .... 3"": .-""'-"2 5’7"; 0 - O 5 % M '- 1 ,v'l‘. I I“). ,Il‘. III-i ’ O . 02 % M -: fl fo A A a: ‘_ 1“] .1 ~3 OJH% M u ‘ lfii if“. A ’2 3 l 0 . 0 0 2 95 M “:3 I [I _ l - J; --. . ___, J: III-II EINUDUAMS‘R- "Sway-p.11 4 .2: 11:49AM Internal size standard (size in nucleotides) I ) Primer peak Wild-type K-ras sequences peak D Mutant K-ras sequences peak Figure 8..A fragment analysis picture of 0.05%, 0.02%, 0.01%, and 0.002% mutant R-ras sequences. All the peaks in different serial dilution samples were aligned by fragment size. (3 Intra-sample variability The capillary electrophoresis followed by RE-PCR was performed as described earlier in serial dilution assay. To determine the intra-sample variability of analytical method, each % mutant sample was analyzed in triplicate and peak area was obtained as shown in Table 7. Figure 9 shows the data range that provided the best linear relationship 67 between log % mutant vs. log mean area. Data on log mutant and log mean area are shown in Table 8. Table 7. Intra-sample variation. % Area 1 Area 2 Area 3 Mean SD CV (%) IMutant Area 100 25677 25515 23167 24786 1405 5.669 75 25415 25667 24712 25265 495 1.959 50 24811 23961 25885 24886 964 3.874 25 23898 23603 21818 23106 1125 4.869 10 18414 18914 15143 17490 2048 11.710 5 12471 14591 11696 12919 1499 11.603 1 6902 6807 6129 6613 422 6.381 0.5 4596 4670 4376 4547 153 3.365 0.25 4110 4179 3111 3800 598 15.737 0.1 1924 1731 1701 1785 121 6.779 0.05 974 999 989 987 13 1.317 0.02 492 478 452 474 20 4.219 0.01 460 397 497 451 51 11.308 0.002 na na 417 417 - — Table 8. Intra-sample variation. Calculation of log % mutant and log mean area of data shown in Table 7. Log % Log Area Log Area Log Area Log Mean SD IMutant 1 2 3 Area 2.000 4.410 4.407 4.365 4.394 0.025 1.875 4.405 4.409 4.393 4.402 0.009 1.699 4.395 4.380 4.413 4.396 0.017 1.398 4.378 4.373 4.339 4.363 0.021 1.000 4.265 4.277 4.180 4.241 0.053 0.699 4.096 4.164 4.068 4.109 0.049 0.000 3.839 3.833 3.787 3.820 0.028 -0.301 3.662 3.669 3.641 3.658 0.015 -0.602 3.614 3.621 3.493 3.576 0.072 -1.000 3.284 3.238 3.231 3.251 0.029 -l.301 2.989 3.000 2.995 2.994 0.006 -l.699 2.692 2.679 2.655 2.676 0.019 -2.000 2.663 2.599 2.696 2.653 0.050 -2.699 na na 2.620 2.620 - 68 l Serial Dilution (Mutantrlildrtype) Assay - Intra-samples , ,lllll, ,- .5 .-l y = 0.4563x + 3.7424 4 {MM 2 R = 0.9673 LogiMean.Area N —1.5 —1 -0 5 0 O 5 1 1 5 2 Log $3Mntant J Figure 9. A plot of log % mutant vs. log mean area (Intra- sample variation). (3 Inter-sample variability Three completely different sets of samples were prepared and analyzed to determine the repeatability of the entire process. The data from this triplicate experiment are shown in Table 9. Figure 10 shows the data range that provided the best linear relationship between log % mutant vs. log mean area. Data on log % mutant and log mean area are shown in Table 10. 69 Table 9. Inter-sample variation. % Area 1 Area 2 Area 3 Mean SD CV (‘15) IMutant Area 100 25677 26186 22591 24818 1945 7.837 75 25415 26389 26338 26047 548 2.104 50 24811 24155 21052 23339 2008 8.604 25 23898 20563 20544 21668 1931 8.912 10 18414 16587 15342 16781 1545 9.207 5 12471 14242 14525 13746 1113 8.097 1 6902 6253 6222 6459 384 5.945 0.5 4596 4557 4342 4498 137 3.046 0.25 4110 4130 3343 3861 449 11.629 0.1 1924 1850 1670 1815 131 7.218 0.05 974 917 909 933 35 3.751 0.02 492 494 409 465 49 10.538 0.01 460 397 492 450 48 10.667 0.002 414 na na 414 - - Table 10. Inter-sample variation. Calculation of log of % mutant and log mean area on data showed in Table 9. Log % Log Area Log Area Log Area Log Mean SD Mutant 1 2 3 Area 2.000 4.410 4.418 4.354 4.394 0.035 1.875 4.405 4.421 4.421 4.416 0.009 1.699 4.395 4.383 4.323 4.367 0.038 1.398 4.378 4.313 4.313 4.335 0.038 1.000 4.265 4.220 4.186 4.224 0.040 0.699 4.096 4.154 4.162 4.137 0.036 0.000 3.839 3.796 3.794 3.810 0.025 -0.301 3.662 3.659 3.638 3.653 0.013 -0.602 3.614 3.616 3.524 3.585 0.052 -1.000 3.284 3.267 3.223 3.258 0.032 -1.301 2.989 2.962 2.959 2.970 0.016 -1.699 2.692 2.694 2.612 2.666 0.047 -2.000 2.663 2.599 2.692 2.651 0.048 -2.699 2.617 Na na 2.617 - 70 Serial Dilution (Mutant:Ni1d-type) assay - Inter-samples I i i .1L~—" -1 -O.5 0 0.5 log % Mutant sample variation). Figure 10..A plot of log % mutant 7] 'PM vs. y = 0.4505x + 3.7356 R2 = 0.9571 log mean area (Inter- DISCUSSION 72 DNA Isolation from Positive and.Negative Controls The concentration of DNA isolated from SW480 cell line (positive control), PBLs (negative control), and III Urine are presented in Table l. The ratio of optical density measured at 260nm and 280nm is greater than 1.8 in all cases, indicating good purity of DNA in the isolated samples. Detection of K-ras Sequences after RE-PCR and Gel Electrophoresis (Qualitative Method) A previously described RE-PCR method was used as a starting mathod URI Y.H. 2004) anmi modified irl order to achieve the goals of this study. 0 First-stage PCR.and First-round of BstNI Digestion First-stage PCR would amplify both mutant and wild- type K-ras sequences to 125 bps as shown in Figure 11 Sense primer creates an artificial BstNI digestion site at codon 11 and 12 in the wild-type K-ras; since mutant K-ras differs from wild-type at codon 12 the sense primer does not create the artificial BstNI digestion site on the mutant K-ras. 73 BstNI restriction enzyme would cut the wild-type K-ras at the BstNI digestion site into 80 and 45 bps fragments. Since, no BstNI digestion site was created in the mutant K- ras, the fragment remains unchanged at 125 bps. However, as observed from Figure l rm) bands were seen for' positive control (SW480), negative control (PBLs), normal urine negative for mutant K-ras either after the first stage PCR or after the first BstNI digestion, this may have resulted from the very low amount of DNA in the samples. 0 Second-stage PCR and Second-round of BstNI Digestion The objective of the study was to qualitatively and quantitatively identify the mutant K-ras. The RE-PCR followed by gel electrophoresis method was developed by Su YH to detect K-ras qualitatively. In order to quantitatively detect both mutant and wild type K-ras sequences using capillary electrophoresis the K-ras sequences had to be labeled with fluorescence. In order to separate wild—type and mutant K-ras on capillary electrophoresis wild-type and mutant K-ras had to be cleaved. differently' during the second. PCR. and 'BstNI. An antisense primer, as shown in Table 1 (Antisense Primer Set II) was specifically developed for this purpose. The antisense primer in the primer set II was labeled with 6- 74 FAM (6—carboxy1 fluorescein) at 5' end. Incorporation of 6- carboxyl fluorescein imparted the fluorescence property needed 1J1 the detection anmi quantification cu? K-ras using the capillary electrophoresis. During second stage PCR; sense primer attaches to the K-ras sequences so that it introduces BstNI (digestion site :u1 the wild-type K-ras, while in mutant K-ras the base sequence is different because of the mutation, therefore, the BstNI digestion site is not created. The gel picture (Figure 2) of SW480, PBL, and normal urine negative for mutant K—ras shows the fragment sizes of 92 bps. This would have resulted from the amplification of mutant or wild-type K—ras as explained by Figure 11. The gel picture of PBLs and normal urine following the second- round of BstNI digestion shows the presence of bands of 62 and 30 bps and no band of 92 bps. While the band for SW480 appear at 92 bps. This could be explained on the basis that sense primer used during second PCR would create an artificial BstNI digestion site at codon 11 and 12 in the wild-type K—ras; since mutant K-ras differs from wild-type at codon 12 the sense primer would not create the artificial BstNI digestion site on the mutant K-ras. During the second BstNI digestion, the BstNI restriction enzyme would cut the wild-type K-ras into 62 and 30 bps fragments. 75 Since, no BstNI digestion site is created; the mutant K-ras fragment remains unchanged at 92 bps. The results show that the RE—PCR method followed by gel electrophoresis was able IX) specifically' detect presence of mutant K-ras in the DNA samples. Ability of Urine Isolation Method to Detect Low Molecular weight Mutant R-ras Sequences Gel picture in Figure 3 shows presence of bands at 62 and 30 bps for PBL and normal urine negative for mutant K- ras. This could be explained as described earlier (under RE-PCR section) and presented in Figure 11. Samples of various amounts of low molecular weight 125 bps SW480 after first PCR mixed with normal urine shows the presence of band at 92 bps. This could be explained as described earlier (under RE-PCR section) and presented in Figure 11. This data indicates that the urine isolation. method. was able to isolate low molecular weight DNA fragments. 76 Serial Dilution Assay o Qualitative Analysis of Mutant R-ras Sequences As observed in Fig. 4 the density of the band for fragment 92 bps correlates qualitatively with the concentration of SW480 in the samples. With the decrease in the density of band for fragment 92 bps as the concentration of SW480 decreases in the sample, as expected, there is a corresponding increase in the density of the bands for fragments 62 and 30 bps obtained by second BstNI digestion of 92 bps band of the wild type K-ras. From the gel picture it is clear that even at the concentration as low as 0.02% of SW480 band at 92 bps is visible. This clearly shows that the qualitative method used to detect K- ras is very sensitive. 0 Quantitative Analysis of Mutant R-ras Sequences in Serial Dilution Assay Quantitative Analysis A typical electrpherogram is shown in figure 6. The fragments were sized. by comparing to G8350-250 internal size standard. The expected sizes for the mutant K-ras sequences and wild-type K-ras sequences were 92 bps and 62 77 bps, respectively. The observed sizes were within 5 bps of expected sizes. This difference may be due to the conformation of the DNA or mobility variation among dyes. A previous research study had reported a similar shift of 5 bps during the quantitative analysis of the FAM-labeled fragment for factor V Leiden by capillary electrophoresis using .ABI Prism. 310 (Benson. J 1999). Another study' had reported a shift iJIJZ bps while quantifying Apolipoprotein using capillary electrophoresis using INN: Prism 310 (Sell S.M. 1997). C) Intra-sample variability Data of intra-sample variability as seen in Table 7 indicates that the analytical method was able to quantify the mutant K-ras consistently as indicated by low standard deviation even in the samples containing mutant K-ras as low as 0.05%. The coefficient of variation (CV) was found to be less than 15%. 0 Figure 9 shows plot of log 6 mutant vs. log mean area. A high value of coefficient of determination (R2) 0.9673, close to 1, indicates a good correlation between log % mutant and log mean area in the range of 50 to 0.05% mutant K-ras. 78 c) Inter-sample variability As seen from the data in Table 9, results obtained from all the three sample set is very consistent (low values of standard deviation) indicating that IXXB sample preparation method is reproducible even for the dilute samples containing mutant K-ras as low as 0.05%. The coefficient of O variation (CV) was found to be less than 15%. 79 CONCLUSIONS 80 ® The QIAamp DNA Mini Kit was used to obtain genomic DNA from PBLs (negative control) and SW480 (positive control). Method to isolate DNA from urine sample was adopted from previous studies and modified to achieve the objectives of these studies. Two stage Restriction Enriched Polymerase Chain Reaction followed by gel electrophoresis was used to detect mutant K—ras sequences without any interference from the wild-type KEras sequences. This method was used as the qualitative method for the detection of mutant K-ras. First-stage PCR amplified both mutant and wild-type K-ras. First-round of BstNI digestion was carried out after first stage RE-PCR with primer set I in order to cut the amplified wild-type K-ras sequences to 85 and 40 bps. However, due to the lower DNA concentration; bands for mutant and wild-type K-ras were not observed in gel electrophoresis following first-stage RE-PCR and first- round of BstNI digestion. Second-stage PCR amplified both mutant and wild—type K-ras. Second-round of BstNI digestion was carried out after second-stage PCR with primer set II. The second-round of BstNI digestion cut the amplified wild- type K-ras sequences in 62 and 30 bps as observed in gel electrophoresis, while IXXE mutant K-ras kxnmi was observed at 92 bps in SW480. Qualitative method was able to detect 8] presence of mutant K-ras in 0.02% SW480 sample, indicating the method is very sensitive. Capillary electrophoresis was used as a quantitative method for analysis of mutant K-ras; quantification was <3 performed using ABI PRISM GeneScan Analysis software. Capillary electrophoresis was atde IX) quantify the mmtant K—ras consistently' even in tflue dilute samples containing mutant K-ras as ihnv as 0.05%. IX good correlation between log % mutant and log mean area was obtained in the range of 50 to 0.05% mutant. The whole process of DNA isolation followed by RE-PCR followed by quantitative analysis of' mutant K-ras using capillary electrophoresis was feund IX) be reproducible as indicated by Ijma low correlation of variation (CV) (<15%) values between the three sets of samples prepared and analyzed at a different time. Also, the intra-sample variability was found to be low as indicated by low CV (<15%). A qualitative and a quantitative method was developed to detect the presence of mutant K-ras in urine sample which could potentially be used in combination with other screening' methods for the early detection of colorectal cancer in patients, which may improve the survival rate in future. Testing' for Imutated. K-ras during follow-up after 82 resection from colorectal cancer may provide new tools for detection of recurrence or second primary tumors and thus may serve IX) identify appropriate treatments or IX) avoid ineffective treatments, and thus may increase survival. 83 APPENDIX 84 Materials 10. Positive Control, Genomic DNA from SW480 cell line (American Type Culture Collection (ATCC)) possessing a mutation in codon 12 of the K-ras proto-oncogene was used as a positive control. Negative Control, Genomic DNA from Peripheral Blood Leucocytes (PBLs) was used as a negative control for K-ras codon 12. Urine Specimen EDTA(ethylenedinitrilo) tetraacetic acid), Disodium Salt, Dihydrate, Crystal, product # 8993-01, Mallinckrodt Baker , Inc., Phillipsburg, NJ. Fifteen mL Conical Polypropylene Screw Cap Tube, part # 430052, Corning Inc., Corning, NY. Guanidine Isothiocyanate, catalog # 15535-016, Invitrogen Corporation, Carlsbad, CA. Fifty mL Conical Polypropylene Screw Cap Tubes, part # 430290, Corning, Inc., Corning, NY. Wizard® Plus Minipreps DNA Purification Systems, catalog # A7100, Promega Corporation, Madison, WI. 1.5 mL macrocentrifuge tube (Eppendrof tubes), catalog # 2236411-1, Brinkmann Instruments Ltd., Canada. SW480 cell line, ATCC® # CCL-228, American Type Culture Collection (ATCC), Manassas, VA. 85 ll. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Sodiumt Chloride, Crystals, product # 3624-05, Mallinckrodt Baker Inc., Phillipsburg, NJ. Potassium Chloride, Crystals, lot # 707202, Columbus Chemical Industries, Inc., Columbus, WI. Sodium Phosphate Monobasic (NaH2PO4), Anhydrous, lot # 126H04925, Sigma-Aldrich, St. Louis, MO. Potassium Phosphate Monobasic (KH2PO4), Batch # 114K0240, Sigma-Aldrich, St. Louis, MO. QIAamp® DNA Mini Kit, catalog # 51304, QIAGEN Inc., Valencia, CA. Ethyl alcohol 200 pmoof (Exhanol), catalog # 111USP200, Pharmaco Products, Brookfield, CT. 0.2 ml thin wall PCR tubes with attached cap, catalog # 501-PCR, Dot Scientific Incorporated, Burton, MI. GeneAmp PCR Buffer 10X, part # N808-0240, Applied Biosystems, Foster city, CA. AmpliTanD Gold DNA polymerase, part # N808-0240, Applied Biosystems, Foster city, CA. dATP, dCTP, dGTP, and dTTP, part # N8080007, Applied Biosystems, Foster city, CA. Primer set I and II (Table l), Macromolecular Structure facility, Michigan State University, East Lansing, MI. 86 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. BstNI Endonuclease Enzyme (source: Bacillus stearothermophilus IQ (EL Comb)), catalog It R0168S, New England Biolabs Inc., Beverly, MA. NEBuffer 2, 10X, catalog # R01685, New England Biolabs Inc., Beverly, MA. Acrylamide, lot # 14328820, Boehringer Mannheim corp., Indianapolis, IN. BIS (N, N’-methylene-bis-acrylamide), catalog # 161- 0201, Bio-Rad Laboratories, Hercules, CA. Trizma®> Base, catalog II T1503-500G, Sigma-Aldrich, St. Louis, MO. Glacial acetic acid, lot # 39076, EM Industries Inc., Gibbstown, NJ. N, N, N’, N'-Tetramethylethylenediamine (TEMED), lot # 14336120, Boehringer Mannheim Corporation, Indianapolis, IN. Ammonium Persulphate, Crystal, lot # C49331, Mallinckrodt Baker, Inc., Phillipsburg, NJ. 5X Nucleic Acid Sample Loading Buffer, catalog # 161-0767, Bio—Rad, Richmond, CA. This loading dye was received. along ‘with. EZ load 100 km) Molecular Ruler, catalog # 170-8352. 50-500 bp Molecular Weight Marker, Ref # 74605-250, PEL-FREEZ, Dynal Biotech, Brown Deer, WI. Ethidium Bromide Solution (10mg/mL), catalog # E1510, Sigma-Aldrich, St. Louis, MO. 87 33. 34. 35. 36. 37. 38. 39. Polaroid® Type 667 Black and White Instant Pack Film, Polaroid Incorporation, Cambridge, MA. Matrix Standard Sets, Applied Biosystems, Foster city, CA. a)Fiuorescent Amidite Matrix Standards Kit (contains one tube each of 6—FAM-, HEX-, TED—, TAMRA-, and ROX-labeled DNA), part # 401546. kn NED Matrix Standard, part # 402996. Formamide, catalog # 11814320001, Roche Diagnostics, Indianapolis, IA. MicroAmp 0.2 mL sample tube, part # N801-0531, Applied Biosystems, Foster city, CA. Performance Optimized Polymer (POP—4rM polymer), part # 402838, Applied Biosystems, Foster city, CA. 10X Genetic Analyzer Buffer with EDTA, part # 402824, Applied Biosystems, Foster city, CA. GeneScanflLBSO ROX® Internal Lane Size Standard, part # 401735, Applied Biosystems, Foster city, CA. 88 Instruments 1. 10. GS Gene Linker UV chamber, serial # B05BR0581, Bio- Rad, Richmond, CA. Precision Water bath—micro, serial # 9503-107, Precision Scientific Inc., Winchester, VA. GeneQuant RNA/DNA. Calculator, Serial II 80-2111-98, Biochrom Ltd, Cambridge Science Park, Cambridge, England. Sorvall TC6 tabletop centrifuge, Serial II 9204183, Sorvall, Thermo Electron Corporation, Asheville, NC. Lab Rotator, Model # 2314, Barnstead International, Dubuque, IA. Sorvall Biofuge® Pico, Serial # 40245700, Sorvall, Thermo Electron Corporation, Asheville, NC. Precision Water Bath, Model II 181, Precision Scientific Inc., Winchester, VA. Vortexer, Type 16700 Mixture, Model # M16715, Barnstead Thermodyne, Dubuque, IA. SpeedVac SC100 System, serial # SC100-OK47405-1A, Savant Instruments, Farmingdale, NY. GeneAmp® PCR System 9700, serial # A965-2050316, Applied Biosystems, Foster city, CA. 89 ll. 12. 13. 14. 15. l6. l7. Precision Shaking Water Bath, serial It 26AT-5, GCA Corporation, Precision Scientific Group, Chicago, IL. Mini-PROTEAN® SKI Cell electrophoresis chamber, serial. # 125BR 36702, aux) a Model 1000/500 Power Supply, Bio-RAD, Richmond, CA. American Rotator V, serial # 030467, American Dade, American Hospital Supply Corporation, Miami, FL. Chromato-Vue Transilluminator, Model # TS-36, UVP Incorporated, San Gabriel, CA. Fotodyne FCR-10 camera, Fotodyne Incorporated, Hartland, WI. ABI PRISM® 310 Genetic Analyzer, Serial # 310- 95080145, Applied Biosystems, Foster city, CA. [NUX Thermal Cycler, serial It P3254, Perkin. Elmer, Norwalk, CT. 90 IMutant and.Wild-type K-ras Sequences during Restriction Enriched Polymerase Chain Reaction 0 Wild-type K—ras exon 1 DNA Sequence 5' ATGACTGAAT ATAAACTTGT GGTAG'ITGGA GCTGGTGGCG TAGGCAAGAG IIIIIIIIII IIIIIIIIII IIIIIIIIII IIIIIIIIIII IIIIIIIIII 3' TACTGACTTA TA'I'ITGAACA CCATCAACCT CGACCACCGC ATCCGTTCTC TGCCTTGACG ATACAGCTAA 'IICAGAATCA I | I IGTGGAC 3’ llllllllll IIIIIIIIII IIIIIIIIII IIIIIIIIII ACGGAACTGC TATGTCGATT AAGTC‘I‘I’AGT AAAACACCTG 5' 0 .Mutant K—ras Sequence Mutant K-ras sequence differs from the wild-type K-ras at the (xxkfll 12 (GGT, highlighted 131 the sequence above) and could have either of following sequence: C) GAT o GTT C) GCT C) AGT 0 TGT C) CGT > Firs t-stage PCR Following Primers were used during the first-stage PCR: 0 Sense Primer Set I 5' GCTCTTCGTGGTGTGGTGTCCATATAAACTTGTGGTAGTTGGACCT 3’ 91 0 Antisense Primer Set I 5' GCTCTTCGTGGTGTGGTGTCCCGTCCACAAAATGATTCTGA 3' Mutant and Wild-type K-ras amplifies to 125 bps during the first stage PCR. Sense jprimer creates an .artificial BstNI digestion site (by encoding G —+ C substitution at the first position of codon 11) at codon 11 and 12 in the wild- type K-ras (highlighted in the sequence below); since mutant K-ras differs from wild-type at CXani 12 the sense primer does not create the artificial BstNI digestion site on the mutant K-ras. 0 Amplified wild-type K~ras after first-stag: PCR 5' GCT C'I'I'CGTGGTG TGGTGTCCAT ATAAACTTGT GGTAG'ITGGA CC GGTGGCG Ill llllllllll Illlllllll IIIIIIIIII IIIIIIIIII IIIIIIIIIII 3' CGA GAAGCACCAC ACCACAGGTA TATTTGAACA CCATCAACCT GGTCACCGC TAGGCAAGAG TGCCTTGACG ATACAGCTAA TICAGAATCA | | I IGTGGAC IIIIIIIIII |||I||I||I ||I|||||Il I|||II||I| ||||||I|I| Arcco'r'rcrc ACGGAACTGC TATGTCGATT AAGTCTTAGT AAAACACCTG GGGACACCAC ACCACGAAGA GC 3' IIIIIIIIII IIIIIIIIII II CCCTGTGGTG TGGTGC‘I‘I’CT CG 5' > First-round of BstNI Digestion BstNI restriction enzyme cuts the Maid-type K-ras at the BstNI digestion site created by sense primer used in the first stage PCR into 80 and 45 bps fragments (shown 92 below). Since, no BstNI digestion site was created in the mutant K-ras, the fragment remains unchanged at 125 bps. o Wild-type K—ras 45 bps fragment following first-round of BstNI digestion 5’ GCT CTTCGTGGTG TGGTGTCCAT ATAAACTTGT GGTAGTTGGA CC III IIIIIIIIII IIIIIIIIII Illlllllll IIIIIIIIII ll 3' CGA GAAGCACCAC ACCACAGGTA TA‘ITTGAACA CCATCAACCT GGA 0 Wild—type K—ras 80 bps fragment following first-round of BstNI digestion TGGTGGCG TAGGCAAGAG TGCCTTGACG ATACAGCTAA TICAGAATCA I I | IGTGGAC IIIIIIII IIIIIIIIII IIIIIIIIII IIIIIIIIII IIIIIIIIII IIIIIIIIII CCACCGC ATCCG'ITCTC ACGGAACTGC TATGTCGAT‘I’ AAGTC‘I‘I’AGT AAAACACCTG GGGACACCAC ACCACGAAGA GC 3’ IIIIIIIIII llllllllll II CCCTGTGGTG TGGTGCTTCT cc 5' 0 Mutant K-ras 125 bps fragments 5’ GCT CTTCGTGGTG TGGTGTCCAT ATAAACTTGT GGTAGTTGGA GCTGGTGGCG Ill IIIIIIIIII IIIIIIIIII IIIIIIIIII IIIIIIIIII Illllllllll 3' CGA GAAGCACCAC ACCACAGGTA TA'ITI'GAACA CCATCAACCT GGACCACCGC TAGGCAAGAG TGCCTTGACG ATACAGCTAA TICAGAATCA I I | IGTGGAC IIIIIIIIII IlIIIIlIIl IllIlIIIII IIIIIIIIII ||||I||||I ATCCGTTCTC ACGGAACTGC TATGTCGAIT AAGTCTI'AGT AAAACACCTG GGGACACCAC ACCACGAAGA GC 3’ IIIIIIIIII llllllllll ll CCCTGTGGTG TGGTGCTTCT co 5’ P Second-stage PCR In the second-stage PCR following sense and antisense primers were used. The antisense primer was labeled with 6- 93 FAM for the quantitative detection of the K-ras using capillary electrophoresis. 0 Sense Primer Set II 5’ ACT GAA TAT AAA CTT GTG GTA G'I‘l' GGA OCT 3’ 0 Antisense Primer Set II 5' more AAG ch ACA AAA TGA TTC TGA ATT AGC 3' Mutant and Wild-type K-ras amplifies to 92 bps during the second stage PCR. The 92 bps sequence of wild-type K- ras is shown below. Sense primer creates an artificial BstNI digestion site at codon 11 and 12 in the wild-type K- ras (highlighted in the sequence below); since mutant K-ras differs from wild-type at codon 12 the sense primer does not create the artificial BstNI digestion site on the mutant K-ras. 0 Wild-type K-ras Fragment size 92 bps after second- stage PCR 5’ ACTGAAT ATAAACTTGT GGTAGTTGGA GCTGGTGGCG TAGGCAAGAG TGCCTTGACG lllllll IIIIIIIIII IIIIIIIIII IIIIIIIIIII IIIIIIIIII IIIIIIIIII 3' TGAC‘I‘I’A TATTTGAACA CCATCAACCT GGACCACCGC Achorrcrc ACGGAACTGC I ATACAGCTAA TICAGAATCA I I I IGTGGAC TTCAG 3’ IIIIIIIIII IIIIIIIIII IIIIIIIIII IIII TATGTCGATI' AAGTCTTAGT AAAACACCTG AAGTC 5' 94 > Second-round of BstNI Digestion BstNI restriction enzyme cuts the Maid-type K-ras at the BstNI digestion site cmeated In; sense primer used in the second stage PCR into 62 anmi 30 bps fragments (shown below). Since, IX) BstNI digestion. site» was created; the mutant K-ras fragment remains unchanged at 92 bps. o Wild-type K-ras 30 bps fragment following second-round of BstNI digestion 5’ ACTGAAT ATAAACTTGT GGTAGTTGGA CC 3’ IIIIIII llllllllll IIIIIIIIII ll 3' TGACTTA TA'ITTGAACA CCATCAACCT GGA 5' 0 Wild-type K—ras 62 bps fragment following second-round of BstNI digestion 5’ TGGTGGCG TAGGCAAGAG TGCCTTGACG ATACAGCTAA TTCAGAATCA I I I IGTGGAC ||III||I II|I|||||| ||||I||III IIIIIIIIII I||||||I|| ||||II|I|| 3’ CCACCGC ATCCG'ITCTC ACGGAACTGC TATGTCGATT AAGTCTTAGT AAAACACCTG TTCAG i? IIIII AAGTCM 5' 0 .Mutant K-ras 92 bps following second-round of BstNI digestion 5’ ACTGAAT ATAAACTTGT GGTAGTTGGA GCTGGTGGCG TAGGCAAGAG TGCCTTGACG lllllll IIIIIIIIII Illlllllll IIIIIIIIII Illlllllll IIIIIIIIII 3' TGACTTA TA‘ITTGAACA CCATCAACCT GGACCACCGC ATCCGTTCTC ACGGAACTGC ATACAGCTAA TTCAGAATCA I I | IGTGGAC TTCAG 3’ IIIIIIIIII IIIIIIIIII IIIIIIIIII IIII TATGTCGATT AAGTCTTAGT AAAACACCTG AAGTC 5' Figure 11. Mutant and‘Nild-type R-ras Sequences during Restriction-Enriched Polymerase Chain Reaction 95 REFERENCS 96 Amado R, Wolf M, Peeters M, et a1. Wild-Type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. Journal of Clinical Oncology. 2008; 10: 1626-1634. American Joint Committee on Cancer. Colon and rectum. Philadelphia: Lippincott—Raven, 2002; 113. Anker P, Lyautey J, Lederrey C, et a1. Circulating nucleic acids in plasma or serum. Clin Chim Acta 2001; 313: 143- 146. Anthony T, Fleming J, Bieligk S, et a1. Postoperative colorectal cancer surveillance. J Am Coll Surg 2000; 190: 737—749. Applied Biosystems https://docs.appliedbiosystems.com/pebiodocs/00903565.pdf Audisio R, Setti—Carraro P, Segela M, et al. Follow-up in colorectal cancer patients: a cost-benefit analysis. Ann Surg Oncol 1996; 3: 349—357. August D.A., Ottow R.T., Sugarbaker P.H. Clinical perspective of human colorectal cancer metastasis. Cancer metastasis Rev. 1984; 3(4): 303-324. Bandipalliam P, Garber J, Kolodner R.D., Syngal S, et al. Clinical presentation correlates with the type of mismatch repair gene involved in hereditary nonpolyposis colon cancer. Gastroenterology 2004; 126: 936—937. Barbacid M. ras genes. Annu. Rev. Biochem. 1987; 56: 779- 827. Benson J, Ellingsen D, Mary A, et al. Multiplex analysis of mutations 1J1 four genes using fluorescence scanning technology. Thrombosis Research. 1999; 96: 57-64. 97 Bhowmick NUA.,Neilson E.G., Moses, H.L. Stromal fibroblasts in cancer initiation and progression. Nature 2004; 432: 332-337. Bischoff F.Z., Nguyen D.D., Marquez-Do D, et al. Noninvasive determination of feta RhD status using fetal DNA in maternal serum and PCR. J Soc Gynecol Investig 1999; 6: 64-69. Boguski M.S. and McCormick F. Proteins regulating Ras and its relatives. Nature 1993; 366: 643-654. Bokemeyer C, Bondarenko I, Hartmann J, et al. KRAS status and efficacy of first-line treatment of patients with metastatic colorectal cancer (mCRC) with FOLFOX with or without cetuximab: The OPUS experience. J Clin Oncol 26: 2008. Boland C.R. and Ricciardiello L. How many mutations does it take to maker a tumor? Proc Natl Acad Sci USA 1999; 96(26): 14675-14677. Bollag M and McCormick F. Regulators and effectors of ras proteins. Annu. Rev. Cell Biol. 1991; 7: 601-632. Bos J.L. Ras oncogenes in human cancer: a review. Cancer Res. 1989; 49:4682-4689. Bos J.L., Fearon E.R., Hamilton S.R., et a1. prevalence of ras gene mutations in human colorectal cancers. Nature 1987; 327: 293-297. Botezatu I, Serdyuk O, Potapova G, et al. Genetic analysis of DNA excreted in urine: a new approach for detecting specific genomic DNA sequences from cells dying in an organism. Clin Chem 2000; 46: 1078-1084. 98 Calvert P.M. and Frucht H. The genetics of colorectal cancer. Ann Intern Med 2002; 137: 603-12. Cancer Statistics, 2005. From website: http://www.cancer.org/downloads/STT/Cancer_Statistics_2005 Cancer Statistics, 2006. From website: http://www.cancer.org/downloads/STT/CAFF2006PWSecured.pdf Chaung D.C. The genetic basis of colorectal cancer: insights into critical pathways of tumorigenesis. Gastroenterology 2000; 119:854-865. Chen X.Q., Bonnefoi H, Diebold-Berger C, et al. Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res 1999; 5: 2297-2303. Chen X.Q., Stroun M, Magnenat J.L., et a1. Microsatellite alterations in plasma DNA of small cell lung cancer patients. Nat Med 1996; 2: 1033-1035. Chiu R.E. and Lo Y.M. The biology and diagnostic applications of fetal DNA and RNA in maternal plasma. Curr Top Dev Biol. 2004; 61: 81-111. Cochrane J.P.S., Williams J.T., Faber R.G., et a1. Value of outpatient follow-up after curative surgery for carcinoma of the large bowel. BMJ 1980; 1: 593—595. Colorectal Cancer In: Strward B.W., Kleihues P, eds. World Cancer Report. Lyon: IARC Press, 2003; 198-202. Compton C.C., Fielding L.P., Burgart L.J., et a1. Prognostic factors in colorectal cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 2000; 124: 979. 99 Crespo P and Leon J. Ras proteins in the control of the cell cycle and cell differentiation. Cell Mol. Life Sci. 2000; 57(11): 1613-1636. Cunningham J.M., Christensen E.R., Rester D.J., et al. Hypermethylation of the hMLHl promoter in colon cancer with microsatellite instability. Cancer Res 1998; 58: 3455-3460. David O.F., JoAnn M.S., Sandy C, et al. The nonhomologous end-joining pathway of DNA repair is required for genomic stability and the suppression of translocations. Proc Natl Acad Sci USA 2000; 97(12): 6630-6633. De la Chapelle A. Microsatellite Instability. N Engl J Med 2003; 349: 209-210. Desch C, Benson A, Smith T, et al. Recommended colorectal cancer surveillance guidelines by the American society of clinical oncology. J Clin Oncol 1999; 17: 1312—1323. Desch C, Benson A, Smith T, et al. Update of American Society of Clinical Oncology colorectal cancer surveillance guidelines. J Clin Oncol 2000; 18: 3586—3588. Esteller M, Sanchez-Cespedes M, Rosell R, et al. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res 1999; 59: 67-70. Fearon E.R. and Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990; 61(5): 759-767. Figueredo A, Rumble R.B., Maroun J, et a1. Follow-up of patients with curatively resected colorectal cancer: a practice guideline. BMC Cancer 2003; 3: 26. I00 Fujita J, Yoshida O, Yuasa Y, et al. Ha-ras oncogenes are activated by somatic alterations in human urinary tracts tumors. Nature 1984; 309: 464-466. Gailani M.R., Stahle-Backdahl M, et al. The role of the human homologue of Drosophila patched in sporadic basal cell carcinomas. Nat Genet 1996; 14(1): 78-81. Goessl C, Heicappel R, Munker R, et al. Microsatellite analysis of plasma DNA from patients with clear renal carcinoma. Cancer Res 1998; 58: 4728-4732. Grady W.M. Genetic testing for high-risk colon cancer patients. Gastoenterology 2003; 124: 1574-1594. Greene F.L., Page D.L., Fleming I.D., et al. (eds). AJCC cancer staging manual, 6th Ed. New York: Springer—Verlag, 2002. Half E.E. and Bresalier R.S. Clinical management of hereditary colorectal cancer syndromes. Curr Opin Gastroenterol 2004; 20: 32—42. Hanahan D and Weinberg R.A. The hallmarks of cancer. Cell 2000; 100(1): 57-70. Harman F.S., Nicol C.J., Marin H.E., et a1. Peroxisome proliferator-activated receptor-delta attenuates colon carcinogenesis. Nat Med. 2004; 10: 481-483. Herman J.G., Umar A, Polyak K, et al. Incidence and functional consequences of hMLHl promoter hypermethylation in colorectal carcinoma. Proc Natl Acad Sci USA 1998; 95: 6870-6875. Hesketh R. The oncogenes handbook. Academic press 1995. Hoeijmakers J.H. Genome maintenance mechanisms for preventing cancer. Nature 2001; 411(6835): 366-374. 101 Howe J.R., Roth S, Ringold J.C., et al. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science 1998; 280(5366): 1086-1088. Huebner R.J. and Todaro G.J. Oncogenes of RNA tumor viruses as determinants of cancer. Proc Natl Acad Sci USA 1969; 64(3): 1087-1094. Infobiogen website: http://www.infobiogen.fr/services/chromcancer/Genes/Genelis te.html Itzkowitz S.H. and Hapraz N. Diagnosis and management of dysplasia in patients with inflammatory bowel disease. Gastroenterology 2004; 126: 1634-1648. Itzkowitz S.H. and Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004; 287: G7—G17. Iwasa Y, F Michor, N.L. Komarova, et al. Population genetics of tumor suppressor genes. J Theor Biol 2005; 233(1): 15-23. Jacoby R.E., Schlack S, Cole C.E., et al. A juvenile polyposis tumor suppressor locus at 10q22 is deleted from nonepithelial cells in the lamina propria. Gastroenterology 1997; 112(4): 1398-1403. Jahr S, Hentze H, Englisch S, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001; 61: 1659-1665. James Trosko and Randall Ruch. Cell-cell communication in carcinogenesis. Frontiers in Bioscience. 1998; 3: d208-236. 102 Jass J.R., Whitehall V.L.J., Young J, Leggett B.A. Emerging concepts in colorectal neoplasia. Gastroenterology 2002; 123: 862-876. Jemal A, Murry T, Samuels A, et al. Cancer statistics, 2003. CA Cancer J Clin 2003; 53: 5-26. Jemal A, Tiwari R.C., Murray T, et al. Cancer statistics 2004. CA Cancer J Clin 2004; 54: 8-29. Jung B, Doctolero R.T., Tajima A, et al. Loss of activin type 2 protein expression in microsatellite unstable colon cancers. Gastroenterology. 2004; 126: 654-659. Kahi C.J. and Rex D.K. Current and future trends in colorectal cancer screening. Cancer Metastasis Rev. 2004; 23: 137-144. Kieveit J and Bruinvels D. Detection of recurrence after surgery for colorectal cancer. Eur J Cancer 1995; 31A: 1222-1225. Kinzler K.W. and Vogelstein B. Lessons from hereditary colorectal cancer. Cell 1996; 87: 159-170. Kinzler K.W. and Vogestein B. Cancer-susceptibiliy gens. Gatekeepers and caretakers. Nature 1997; 386(6627): 761- 763. Kjeldsen B.J., Kronborg O, Fenger C, Jorgensen O.D. A prospective randomized study of follow-up after radical surgery for colorectal cancer. Br J Surg 1997; 84: 666-669. Kondo Y and Issa J.P. Epigenetic changes in colorectal cancer. Cancer Metastasis Rev. 2004; 23: 29-39. 103 Kopreski M, Benko F, Kwee C, et al. Detection of mutant K- ras DNA in plasma or serum of patients with colorectal cancer. Br J Cancer 1997; 76: 1293-1299. Kressner U, Bjorheim J, Westring S, et al. Ki-ras mutations and prognosis in colorectal cancer. Eur J Cancer 1998; 43: 518-521. Krok K.L., Lichtenstein G.R. Colorectal cancer in inflammatory bowel disease. Curr Opin Gastoenterol 2004; 20: 43-48. Lengauer C, Kinzler K.W., Vogelstein B. Genetic instabilities in human cancers. Nature 1998; 396(6712): 643-649. Leon J, Guerrero I, Pellicer A. Differential expression of the ras gene family in mice. Mol. Cell Biol. 1987; 7: 1535- 1540. Lichtenstein A.V., Melkonyan H.S., Tomei L.D., et al. Circulating nucleic acids and apoptosis. Ann. N.Y. Acad. Sci. 2001; 945: 239-249. Lo Y.M., Corbetta N, Chamberlain P.F., et a1. Presence of fetal DNA in maternal plasma and serum. Lancet 1997; 350: 485-487. Lowy D.R., Willumsen B.M. Function and regulation of Ras. Annu Rev Biochem 1993; 62: 851-891. Luebeck E.G. and Moolgavkar S.H. Multistage carcinogenesis and the incidence of colorectal cancer. Proc Natl Acad Sci USA 2002; 99(23): 15095-15100. Lynch H.T. and de la Chapelle A. Hereditary colorectal cancer. N Engl J Med 2003; 348: 919-932. 104 Lynch J.P. and Hoops T.C. The genetic pathogenesis of colorectal cancer. Hematol Oncol Clin North Am. 2002; 16: 775-810. Macaluso M, Russo G, Cinti C, et al. Ras family genes: an interesting link between cell cycle and cancer. J. Cell. Physiol. 2002; 192: 125-130. Malumbres M and Pellicer A. RAS pathways to cell cycle control and cell transformation. Fronts. Biosci. 1998; 3: 887-912. McCormick F. Signaling networks that cause cancer. Trends Cell Biol. 1999; 9: M53-M56. Michor F, Iwasa Y, Nowak M.A. Dynamics of cancer progression. Nat Rev Cancer 2004; 4(3):197-205. Milde A, Haas-Rochholz H, Kaatsch HJ. Improved DNA typing of human urine by adding EDTA. Int J Legal Med. 1999; 112: 209-210. Monifar F, Yan Gao M, Arnould L, et al. Concurrent and independent genetic alterations in the stromal and epithelial cells of mammary carcinoma: implications for tumorigenesis. Cancer Res 2000; 60(9): 2562-2596. Mulcahy H.E., Lyautey J, Lederrey C, et al. A prospective study of K-ras mutations in plasma of pancreatic cancer patients. Clin Cancer Res 1998; 4: 271-275. Muller A, et al. MSI-testing in hereditary non-polyposis colorectal carcinoma (HNPCC). Dis Markers 2004; 20(4-5): 225-236. Munger K. Disruption of oncogene/tumor suppressor networks during human carcinogenesis. Cancer Invest 2002; 20(1): 71- 81. 105 Nawroz H, Koch W, Anker P, et al. Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med 1996; 2: 1035-1037. Nowell P.C. The clonal evaluation of tumor cell populations. Science. 1976; 194: 23-28. Nowell P.C. Tumor progression: a brief historic perspective. Semin Cancer Biol. 2002; 12: 261-266. Pells S, Divjak M, Romanowski P, et al. Developmentally- regulated expression of murine K-ras isoforms. Oncogene 1997; 15: 1781-1786. Ponder B.A. Cancer genetics. Nature 2001; 411(6835): 336- 341. Rajagopalan H, Jallepalli P.V., Rago C, et al. Inactivation of hCDC4 can cause chromosomal instability. Nature 2004; 428: 77—81. Rajagopalan H, Nowak M.A., Vogelstein B, et al. The significance of unstable chromosomes in colorectal cancer. Nat Rev Cancer 2003; 3(9): 695-701. Rao M, Yang W, Seifalian A.M., Winslet M.C. Role of cyclooxygenase-2 in the angiogenesis of colorectal cancer. Int J Colorectal Dis. 2004; 19: 1-11. Rodenhuis S and Slebos R.J. Clinical significance of ras oncogene activation in human lung cancer. Cancer Res 1992; 52: 26653-26693. Russo M.W., Wei J.T., Thiny M.T., et al. Digestive and liver disease statistics, 2004. Gastroentrology 2004; 126: 1448-1453. 106 Samuels A, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004; 304: 554. Schoemaker D, Black R, Giles L, Toouli J. Yearly colonoscopy, liver CT and chestradiography do not influence 5-year survival of colorectal cancer patients. Gastroenterology 1998; 114: 7-14. SEER data 1975-2001. From website: http://seer.cancer.gov/csr/1975_2001/results_merged/sect_06 _colon_rectum.pdf Sell S.M. and Ren K. Automated capillary electrophoresis in the genotyping of Apolipoprotein E. Genomics. 1997; 46: 163-164. Sherr C.J. Principles of tumor suppression. Cell 2004; 116(2): 235—246. Sieber O.M., Lipton L, Crabtree M, et al. Multiple colorectal adenomas, classic adenomatous polyposis, and germ-line mutations in MYH. N Engl J Med 2003; 348: 791- 799. Sinicrope F.A. and Gill S. Role of cyclooxygenase-2 in colorectal cancer. Cancer Metastasis Rev. 2004; 23: 63-75. Slebos RJ, Hruban R.H., Dalesio O, et al. Relationship between K-ras oncogenes activation and smoking in adenocarcinoma of the human lung. J Natl Cancer Inst 1991; 83: 1024-1027. Soetikno R.M., Kahng L.S., Ono A, Fujii T. Flat and depressed colorectal neoplasms. Curr Opin Gastroenterol. 2003; 19: 69-75. 107 Su Y.H., Wang M, Brenner D.E., et al. Human Urine contains small, 150 to 250 nucleotide-sized, soluble DNA derived from the circulation and may be useful in the detection of colorectal cancer. J Mol Diagn. 2004; 6(2): 101-107. Van Cutsem E, Lang I, D'haens G, et al. KRAS status and efficacy in the first-line treatment of patients with metastatic colorectal cancer (mCRC) treated with FOLFIRI with or without cetuximab: The CRYSTAL experience. J Clin Oncol 26: 2008. Vasen H.E., Watson P, Mecklin J.P., Lynch H.T. New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the international collaborative group on HNPCC. Gastroenterology 1999; 116: 1453-1456. Veigl M.L., Kasturi L, Olechnowicz J, et al. Biallelic inactivation of hMLHl by epigenetic gene silencing, a novel mechanism causing human MSI cancers. Proc Natl Acad Sci USA 1998; 95: 8698-8702. Venesio T, Molatore S, Cattaneo F, et al. High frequency of MYH gene mutations in a subset of patients with familial adenomatous polyposis. Gastroenterology 2004; 126: 1681- 1685. Vernava A, Longo W, Virgo K, et al. Current follow-up strategies after resection of colon cancer. Results of a survey of members of the American Society of Colon and Rectal Surgeons. Dis Colon Rectum 1994; 37: 573—583. Visvanathan K.V., Pocock R.D., Summerhayes I.C. Preferential and novel activation of H-ras in human bladder carcinomas. Oncogene Res. 1988; 3: 77-86. Vogelstein B and Kinzler K.W. The multistep nature of cancer. Trends Genet 1993; 9(4): 138-141. 108 Vogelstein B, Fearon E.R., Hamilton S.R., et al. Genetic alterations during colorectal-tumor development. N Engl J Med 1988; 319: 525-532. Vogestein B and Kinzler K.W. Cancer genes and the pathways they control. Nat Med 2004; 10(8): 789-799. Wang Z, Shen D, Parsons D.W., et a1. Mutational analysis of the tyrosine phosphatome in colorectal cancers. Science 2004; 304: 1164-1166. Weaver Z, Montagna C, Xu X, et al. Mammary tumors in mice conditionally mutant for Brcal exhibit gross genomic instability and centrosome amplification yet display a recurring distribution of genomic imbalances that is similar to human breast cancer. Oncogene 2002; 21(33): 5097-5107. Winawer S.J., Fletcher R, Rex D.K., et al. Colorectal cancer screening and surveillance: clinical guidelines and rationale-update based on new evidence. Gastroenterology 2003; 124: 544-560. Winawer S.J., Zauber A.G., Ho M.N., et al. Prevention of colorectal cancer by colonoscopic polypectomy. The National Polyp Study Workgroup. N Engl J Med. 1993; 329: 1977-1981. Wong IHN, Lo YMD, Zhang J, Liew C-T, et al. Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res 1999; 59: 71-73. Wyllie A.H. Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature 1980; 284: 555-556. 109 STATE UNIVERSITY LIBRARIES Ill 1111 | 93 03062 5630 “111111)