Download Minimal Residual Disease Quantification Using Consensus Primers

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
1
2
3
4
5
6
7
8
9
10
11
12
Minimal Residual Disease Quantification Using Consensus Primers and HighThroughput IGH Sequencing Universally Predicts Post-Transplant Relapse in
Chronic Lymphocytic Leukemia
13
PBMC were generated from 10 mL of whole blood by centrifugation on a Ficoll-Hypaque
14
gradient, followed by washing with phosphate buffered saline (PBS) and
15
cryopreservation in freezing medium consisting of 50% fetal bovine serum, 10%
16
dimethylsulfoxide (DMSO), and 40% Roswell Park Medical Institute (RPMI) medium.
17
Cells were stored in liquid nitrogen vapor until used for MRD quantification. To assess
18
MRD, samples were removed from cryopreservation, thawed rapidly in a 37°C water
19
bath and washed twice with PBS. A cell lysate was made immediately using Buffer AL
20
with proteinase K from a Qiagen Blood and Tissue kit (Qiagen, Valencia, CA) and the
21
remainder of DNA isolation proceeded according to manufacturer’s instructions. For
22
some samples, DNA was harvested using Gentra Puregene Blood kit (Qiagen) from
23
peripheral blood collected in EDTA tubes per manufacturer’s instructions.
Aaron C. Logan,1 Bing Zhang,2 Balasubramanian Narasimhan,3 Malek Faham,4 Victoria
Carlton,4 Jianbiao Zheng,4 Martin Moorhead,4 Mark R. Krampf,1 Carol D. Jones,2 Amna
N. Waqar,2 James L. Zehnder,2 and David B Miklos1
Supplemental Methods
MRD samples
24
25
IGH amplification and sequencing
26
First stage primers for multiplex PCR were designed to amplify all known
27
germline IGH sequences. Each IGHV segment is amplified by 3 primers, decreasing the
28
likelihood of somatic hypermutations preventing amplification. Primers were optimized
29
such that each possible IGHV and IGHJ segment was amplified at a similar rate so as to
30
minimally skew the repertoire frequency distribution during linear amplification. A given
31
sequence may have been amplified by multiple primers and this was handled
32
bioinformatically during IGH clonotype quantification such that one amplimer was used
33
for quantification of each specific clonotype. This methodology led to slightly different
34
primer designs than have been published previously for similar IGH amplification
35
approaches.1 The numbers of primers and the positions of these primers are shown in
36
Faham and Willis.2
37
At the 5’ ends of IGHV segment primers a universal sequence complementary to
38
a set of second stage PCR primers was appended. Similarly the primers on the IGHJ
39
side had a 5’ tail with a universal sequence complementary to second stage PCR
40
primers. Second stage PCR primers additionally contained a sequence primer site and
41
the P5 sequence used for cluster formation in the Illumina Genome Analyzer sequencer.
42
The primers on the IGHV side of the amplification constituted one of a set of primers,
43
each of which had a 3’ region that annealed to the overhang sequence appended in the
44
first reaction but which further contained one of multiple 6 or 9 base pair indices that
45
allowed for sample multiplexing on the sequencer. Each of these primers further
46
contained a 5’ tail with the P7 sequence used for cluster formation in the Illumina
47
Genome Analyzer sequencer.
48
First stage PCR was carried out using a high fidelity polymerase (AccuPrime, Life
49
Technologies) for 16 cycles. 1/100 of this amplification reaction was then used as the
50
template for a second PCR reaction using the second stage primers that append sample
51
indices and cluster formation sequences. A second stage PCR was carried out for 22
52
cycles. Different samples were pooled for sequencing in the same Illumina Genome
53
Analyzer sequencing lane. The pool was then purified using the QIAquick PCR
54
purification kit (Qiagen).
55
Cluster formation and sequencing was carried out per the manufacturer protocol
56
(Illumina, Inc., La Jolla, CA). Specifically, three sequencing reactions were performed.
57
First 115 bp were sequenced from the IGHJ side sufficient to sequence through the
58
CDR3 junctional sequence from IGH J-to-V. At this point, the synthesized strand was
59
denatured and washed off. A second sequencing primer was annealed that allowed the
60
sample index to be sequenced for 6 cycles to identify the sample. At this point the
61
reverse complement strand was generated in a third sequencing reaction per the
62
Illumina protocol. The final sequencing read of 95 bp obtained from the IGH V-to-J
63
direction provided ample sequence to map the IGHV segment accurately using germline
64
sequences published by the International Immunogenetics (IMGT) Information System.3
65
66
Clonotype determination
67
Algorithmic methods were utilized for clonotype determination. Briefly, sequence data
68
were analyzed to determine the clonotype sequences including mapping to germline V
69
and J consensus sequences.3 First, the forward sequence read was used to map the J
70
segment. After J segment identification, V segments were mapped using the reverse
71
sequence read. The IGHV primer was mapped and the bases under this primer were
72
excluded from further analysis of the reverse read. Thereafter, the next ~70 bases of the
73
reverse read were mapped to the known IGHV segments. Read pairs that did not map to
74
V segments were excluded. The next step in mapping involved identifying the frame that
75
related the forward and reverse reads and this allowed a continuous sequence from J to
76
V to be constructed.
77
To generate a clonotype, identification of at least two identical sequences was
78
required. We developed an algorithm to determine whether similar sequencing reads are
79
the result of biological differences in the initial sample or technical artifact (i.e.,
80
sequencing or PCR error). The algorithm takes into account the number of sequencing
81
reads and the degree of sequence variation between the clonotypes in question. For
82
example, two sequences with one base difference but present at vastly different
83
frequencies were consistent with sequencing or PCR error. On the other hand two
84
sequences with two base differences and present at similar magnitudes were unlikely to
85
arise from sequencing error. Non-functional rearrangements (generally less than 20% of
86
all VDJ rearrangements) are included in the analysis.
87
88
MRD quantification
89
To determine the absolute measure of the total leukemia-derived molecules present in
90
the follow-up sample, we added a known quantity of reference IGH sequence into the
91
reaction and counted the associated sequencing reads. The known quantity of reference
92
IGH sequence was derived from a pool of plasmids containing 3 unique IGH clonotypes,
93
quantified using standard RT-PCR methods. The resulting factor (number of molecules
94
per sequence read) was then applied to the leukemia associated clonal rearrangement
95
reads to obtain an absolute measure of the total leukemia-derived molecules in the
96
reaction. A similar calculation was performed to assess the total number of rearranged
97
IGH molecules, or B-lineage cells, in the reaction. Finally, we calculated the total
98
leukocytes in the reaction by measuring the total DNA in the reaction using standard
99
picogreen methods and RT-PCR using β actin DNA, assuming an average human
100
diploid genome mass of 6.49 picograms. These metrics were combined to calculate a
101
final MRD measurement, which is the number of leukemia-derived molecules divided by
102
the total leukocytes in the sample (capped at 1 million CLL clonotypes per 1 million input
103
PBMC genomes).
104
105
IGH allele-specific oligonucleotide PCR
106
An IGH V-region consensus Taqman probe was first evaluated for predicted success
107
based on a set of criteria including the number, type and position of mismatches.4 If the
108
consensus probe was predicted to be successful, allele-specific primers were designed
109
to work with the probe, with the forward primer annealing 5’ of the probe and the reverse
110
primer annealing in the complementarity determining region 3 (CDR3) region. If the
111
consensus probe was predicted to be unsuccessful or deemed empirically to be
112
insensitive for a specific patient, a CDR3-specific probe and corresponding primers were
113
designed. Q-PCR reactions were performed on an ABI 7900HT real-time PCR
114
instrument (Applied Biosystems, Carlsbad, CA) with 500ng of total leukocyte DNA and
115
Taqman Universal PCR master mix (Applied Biosystems). Human genomic DNA (Roche
116
Diagnostics, Germany) was used as a reference GAPDH standard and the IGH data
117
were normalized to the corresponding GAPDH quantification and the final result was
118
reported as number of CLL IGH copies/μg of human DNA.
119
120
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
140
141
142
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
Supplemental Tables
Supplemental Table 1. Patient characteristics and outcomes
[Separate file due to landscape orientation.]
Supplemental Figures
Supplemental Figure 1. Patient outcomes. Overall (OS) and disease-free survival
(DFS) for the patient cohort studied here are shown.
Supplemental Figure 2. MRD quantification in relapsed and non-relapsed patients.
MRD quantification is shown for patients relapsing within 12 months post-HCT (A) after
12 months post-HCT (B). MRD patterns for patients who remained free of relapse are
shown in (C).signifies a patient (SPN 3723) with apparent MRD progression, but
who died from complications of chronic GVHD prior to meeting criteria for clinical
relapse.
Supplemental References
1.
van Dongen JJ, Langerak AW, Bruggemann M, Evans PA, Hummel M, Lavender
FL et al. Design and standardization of PCR primers and protocols for detection
of clonal immunoglobulin and T-cell receptor gene recombinations in suspect
lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT983936. Leukemia 2003; 17(12): 2257-317.
2.
Faham M, Willis TD. Monitoring health and disease status using clonotype
profiles. In: USPTO ed. Vol. 2011/0207134A1. United States; 2011.
3.
Giudicelli V, Chaume D, Lefranc MP. IMGT/GENE-DB: a comprehensive database
for human and mouse immunoglobulin and T cell receptor genes. Nucleic Acids
Res. 2005;33(Database issue):D256-261.
4.
Ladetto M, Donovan JW, Harig S, Trojan A, Poor C, Schlossnan R et al. Real-Time
polymerase chain reaction of immunoglobulin rearrangements for quantitative
evaluation of minimal residual disease in multiple myeloma. Biol Blood Marrow
Transplant 2000; 6(3): 241-53.