ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 4, pp. 637-652 © Pleiades Publishing, Ltd., 2024.
637
Modification of the Hi-C Technology
for Molecular Genetic Analysis of Formalin-Fixed
Paraffin-Embedded Sections of Tumor Tissues
Maria M. Gridina
1,2,a
*, Yana K. Stepanchuk
1,2
, Miroslav A. Nurridinov
1,2
,
Timofey A. Lagunov
1,2
, Nikita Yu. Torgunakov
1,2
, Artem A. Shadsky
1,2
,
Anastasia I. Ryabova
3
, Nikolay V. Vasiliev
3
, Sergey V. Vtorushin
3,5
,
Tatyana S. Gerashchenko
3
, Evgeny V. Denisov
3
, Mikhail A. Travin
4
,
Maxim A. Korolev
4
, and Veniamin S. Fishman
1,2
1
Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences,
630090 Novosibirsk, Russia
2
Novosibirsk State University, 630090 Novosibirsk, Russia
3
Research Institute of Oncology, Tomsk National Research Medical Center, Russian Academy of Sciences,
634009 Tomsk, Russia
4
Research Institute of Clinical and Experimental Lymphology, Institute of Cytology and Genetics,
Siberian Branch of the Russian Academy of Sciences, 630117 Novosibirsk, Russia
5
Siberian State Medical University, Ministry of Health of Russia, 634050 Tomsk, Russia
a
e-mail: gridinam@gmail.com
Received September 5, 2023
Revised October 31, 2023
Accepted October 31, 2023
AbstractMolecular genetic analysis of tumor tissues is the most important step towards understanding the
mechanisms of cancer development; it is also necessary for the choice of targeted therapy. The Hi-C (high-through-
put chromatin conformation capture) technology can be used to detect various types of genomic variants, includ-
ing balanced chromosomal rearrangements, such as inversions and translocations. We propose a modification
of the Hi-C method for the analysis of chromatin contacts in formalin-fixed paraffin-embedded (FFPE) sections
oftumor tissues. The developed protocol allows to generate high-quality Hi-C data and detect all types of chro-
mosomal rearrangements. We have analyzed various databases to compile a comprehensive list of translocations
that hold clinical importance for the targeted therapy selection. The practical value of molecular genetic testing is
its ability to influence the treatment strategies and to provide prognostic insights. Detecting specific chromosomal
rearrangements can guide the choice of the targeted therapies, which is a critical aspect of personalized medicine
in oncology.
DOI: 10.1134/S0006297924040047
Keywords: chromosomal rearrangements, three-dimensional nuclear organization, oncology, FFPE sections
* To whom correspondence should be addressed.
INTRODUCTION
Together with single nucleotide variants (SNVs),
chromosomal rearrangements, including balanced trans-
locations and inversions, play a key role in the patho-
genesis of various cancers. Current genomic diagnostic
approaches enable genome-wide detection of SNVs and
copy number variations, offering significant insights
into oncogenic processes. However, efficient detection
of balanced chromosomal rearrangements remains
elusive. At the same time, these chromosomal rear-
rangements have been found in almost all types of can-
cer. Moreover, for some tumors, detection of balanced
chromosomal rearrangements is critical for the diag-
nosis, clarification of prognosis, and choice of therapy.
GRIDINA et al.638
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
In many tumors, chromosomal rearrangements
not only accompany the process of tumor develop-
ment, but act as the main cause (driver) of cell onco-
logical transformation. One of the examples is recipro-
cal translocations observed in Burkitt’s lymphoma, in
which the translocation of the MYC gene from chromo-
some 8 to chromosome 14 under the influence of the
immunoglobulin heavy chain enhancer results in dys-
regulation of its expression [1]. If breakpoints occur
within the genes, this can lead to the gene fusions re-
sulting in the formation of chimeric proteins. Such fu-
sion proteins often involve transcription factors (ERG,
MYB) or protein kinases (ABL1, ALK, BRAF, EGFR, JAK2,
RET) that play a pivotal role in the oncogenic process.
The TMPRSS2-ERG gene fusion, which is prevalent in
a majority of prostate adenocarcinomas and approxi-
mately 20% of high-grade prostate intraepithelial neo-
plasias, illustrates this mechanism. TMPRSS2 is a serine
protease regulated by the androgen-dependent promot-
er and its fusion with the ERG oncogene results in the
ERG overexpression, a key event in the prostate cancer
pathogenesis [2]. Similar mechanisms involving ERG
fusion with other partners, such as NDRG1, EWS, and
FUS, have been implicated in other cancer types[3-5].
Gene fusion, a hallmark of various cancers, can
dysregulate gene expression and alter the function of
the encoded protein. Thus, if gene fusion results in the
truncation of one of the fusion partners, this can lead
to its overexpression due to the loss of negative reg-
ulatory elements (e.g., binding sites for microRNA) or
domains determining the protein lifespan. A notable
example is the MYB-NFIB gene fusion in adenoid cys-
tic carcinoma, resulting from the t(6;9) translocation
[6]. In this fusion, the chimeric transcript partially or
completely loses a region encoding the C-terminal reg-
ulatory domain of MYB containing the sites for protein
post-translational modification, as well as a non-cod-
ing sequence essential for the binding of microRNAs.
Consequently, the absence of these regulatory ele-
ments in the MYB portion of the fusion protein leads
to the upregulation of MYB expression and prolonged
protein lifespan [7].
Gene fusions can also lead to the production of
chimeric proteins with significantly altered functional
domains. In the norm, the FGFR3 receptor tyrosine ki-
nase is activated through the homo/heterodimerization
in the presence of fibroblast growth factor (FGF) as a
ligand [8]. The translocation between chromosomes
4 and 7 results in the FGFR3 fusion with BAIAP2L1.
Theresulting chimeric protein possesses the ability for
constitutive, ligand-independent homodimerization.
This aberrant dimerization is facilitated by the BAR
domains of BAIAP2L1, resulting in the FGFR3 kinase
activation and potent oncogenic activity [9].
For the diagnostic purposes and long-term stor-
age, tumor samples are preserved as formalin-fixed
paraffin-embedded (FFPE) tissue blocks through for-
malin fixation and subsequent embedding in paraffin.
FFPE blocks have many advantages, including stability
at room temperature, extended shelf life, and compat-
ibility with immunohistochemical analysis. However,
such fixation and storage of samples can lead to the
degradation of nucleic acids and appearance of arti-
facts, which requires optimization of molecular anal-
ysis methods [10]. Furthermore, the degradation and
modification of nucleic acids in FFPE samples compli-
cate the use of RNA sequencing for the detection of
biomarkers [11].
Routine methods for identification of chromosom-
al rearrangements in tumor tissues include FISH (flu-
orescence in situ hybridization), immunohistochemical
analysis, and RT-PCR. These approaches have obvious
limitations in the detection of novel or complex chro-
mosome rearrangements. Recent advances in high-
throughput sequencing have revolutionized clinical
genetics. Whole-genome sequencing (WGS) and whole-
exome sequencing (WES) using the short-read technol-
ogy have excelled in identifying SNVs and unbalanced
chromosomal rearrangements, but their accuracy in
repetitive genome regions is limited. Detection of bal-
anced rearrangements using WGS and WES depends
on the presence of chimeric reads encompassing the
rearrangement breakpoints and therefore requires a
high sequencing depth. Long-read sequencing methods
(PacBio and Oxford Nanopore) are effective for detect-
ing balanced chromosomal rearrangements, but their
efficiency diminishes when analyzing FFPE samples
due to the DNA degradation. Balanced chromosomal
rearrangements often trigger carcinogenesis through
two mechanisms: gene fusion and disruption of gene
expression resulting from alterations in the gene reg-
ulatory environment. Consequently, RNA sequencing
has emerged as an important tool for analyzing tumor
samples [12-14]. However, this technique demands a
high RNA quality, which is challenging when RNA is
isolated from FFPE samples [11-16]. Degraded RNA
fragments may lack crucial information on the fusion
sites. Moreover, RNA-seq technology faces sensitivity
issues in the case of low expression of fusion tran-
scripts [13] and fusions with non-coding regions [17]
and requires significant sequencing depth (20-30 mil-
lion paired-end reads) or targeted gene enrichment
[11]. The Hi-C (high-throughput chromatin conforma-
tion capture) method has been increasingly used in
recent years as an alternative approach for detect-
ing various types of chromosomal rearrangements.
Theadvantage of the method is its ability to detect bal-
anced rearrangements at a lower sequencing depth.
This efficiency is partly due to the fact that Hi-C does
not rely solely on the reads containing the breakpoint.
Instead, it analyzes changes in the chromatin contact
frequency within broad genomic regions and, therefore,
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 639
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
requires less sequencing depth for the detection of re-
arrangements [18-27].
Here, we propose a new Hi-C protocol for analyz-
ing material from FFPE tumor sections. We introduced
significant modifications to the existing protocols
[28, 29], resulting in a highly reproducible technique
capable of generating high-quality Hi-C data and de-
tecting all types of chromosomal rearrangements. A key
innovation in our approach that distinguishes it from
traditional 3C methods, is the use of a sequence-ag-
nostic nuclease, which not only facilitates detection
of chromosomal rearrangements but also expands the
application of this method to identification of SNVs in
clinically significant loci. We have analyzed various
databases to compile a comprehensive list of translo-
cations that hold clinical importance for selection of a
targeted therapy. The results of modeling conducted in
our study demonstrate that our method has a substan-
tial promise for clinical application.
MATERIALS AND METHODS
Analyzed samples. FFPE sections were obtained
from patients treated at several medical centers of the
Russian Federation. Eight patients were from the On-
cology Research Institute of the Tomsk National Med-
ical Research Center. Three patients (age, 43.6 ± 8.62
years) had morphologically verified grade 4 (G4) brain
tumors (glioblastoma, giant cell glioblastoma, and
diffuse astrocytoma). Five patients (age range, 28-65
years; average age, 50.4 ± 12.9 years) had morpholog-
ically confirmed chondrosarcomas of different lo-
calization (humerus, femur, tibia, pelvic bones, and
sternum); the tumor grades ranged from G2 to G3. Six
patients were treated at the Kemerovo Regional Clin-
ical Hospital; three of them had chronic lymphocytic
leukemia (CLL) and three patients had large cell lym-
phoma (LCL). The diagnoses were established based
on pathomorphological studies of excisional lymph
node biopsies and immunohistochemical verification
using a specialized antibody panel.
Tumor tissue samples were collected during surgi-
cal procedures. The samples were fixed in 10% neutral
buffered formalin for 24 h and embedded in paraffin
using standard techniques. For each tumor specimen,
10μm thick sections were prepared.
FFPE Hi-C. The developed protocol was based on
our previously proposed S1 Hi-C method [30] and in-
cluded the following steps:
1. Deparaffinization:
1.1. An FFPE section was placed in a 1.5-ml tube and
1 ml of lysis buffer Y (150mM Tris pH8.0; 140mM
NaCl, 0.5% Igepal, 1% Triton X-100) was added.
1.2. The FFPE section was incubated at 80°C
for3min.
1.3. Centrifugation was performed at 2500g for 5min.
1.4. The paraffin layer was removed from the solu-
tion surface.
1.5. Steps 1.2.-1.4. were repeated (i.e., the total num-
ber of incubations was two).
2. Lysis:
2.1. After the second centrifugation, the superna-
tant was removed and the pellet was resuspended
in 1ml of lysis buffer H (10mM Tris pH8.0, 10mM
NaCl, 1% Triton X-100, 0.1% SDC, 20% EtOH).
2.2. The sample was incubated at 45°C overnight.
2.3. Centrifugation was performed at 2500g for
5min.
2.4. The precipitate was washed once with 1ml of
lysis buffer Y.
2.5. The sample was incubated in 1ml of lysis buf-
fer Y for 1 h at room temperature on an orbital
shaker.
2.6. Centrifugation was performed at 2500g for
5min.
2.7. The supernatant was removed, and the pel-
let was resuspended in 500 μl of lysis buffer D
(50mM Tris pH 7.5, 0.5mM CaCl
2
, 0.3% SDS).
2.8. The sample was incubated at 37°C for 1h.
2.9. SDS was quenched by adding 91μl of 10% Tri-
ton X-100 for 10min at room temperature.
2.10. Centrifugation was performed at 2500g for
5min.
2.11. The pellet was washed once with 500 μl of
1× S1 nuclease buffer (Thermo Scientific) contain-
ing 1% Triton X-100.
3. Chromatin fragmentation:
3.1. The pellet was resuspended in 80 μl of 1× S1
nuclease buffer.
3.2. 200 U of S1 nuclease (Thermo Scientific) was
added and the mixture was incubated at 37°C
for1h.
3.3. The reaction was stopped by adding 5 μl
of 500 mM EDTA and purified with 1 volume of
AMPure magnetic beads according to the manu-
facturers recommendations.
3.4. Chromatin associated with magnetic beads
was resuspended in 100μl of H
2
O (chromatin re-
mained bound to the beads until the DNA isola-
tion stage).
4. Further steps, including biotin labeling, ligation,
DNA isolation, ligation fragment enrichment, and
preparation of NGS libraries, were performed ac-
cording to the protocol described by Gridina etal.
[31]. DNA quantity was determined with a Qubit
dsDNA HS Assay Kit.
The prepared libraries were sequenced using the
BGI sequencing platform with 150-bp paired-end reads.
The sequencing depth was 10-100 thousand reads per
sample for shallow sequencing and ~80 million paired-
end reads for deep sequencing.
GRIDINA et al.640
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Modeling of chromosomal rearrangements was
performed with the Charm software (https://github.com/
genomech/Charm/) using the published results of ge-
nome-wide Hi-C studies in IMR90 cells [32] (identifiers
SRR1658675, SRR1658676, SRR1658679) to create a da-
tabase of reference contacts. Each chromosomal rear-
rangement was modeled as heterozygous, with a total
number of ~30 million Hi-C contacts. The coordinates
for the boundaries of modeled chromosomal rear-
rangements (Online Resource 1) were rounded to the
nearest 5kb. Rearrangements smaller than 25kb were
scaled up to this threshold.
Construction of contact maps and analysis of
quality metrics of Hi-C libraries. Analysis of Hi-C
data and construction of Hi-C heatmaps were conduct-
ed as outlined in [31]. A modified version of the Juicer
software version 1.6 (available on GitHub: https://
github.com/genomech/juicer1.6_compact) was used to
calculate the quality scores.
RESULTS
Development of Hi-C protocol for FFPE samples
using S1 nuclease. The majority of published Hi-C
protocols have been designed for living cells or fresh
tissues [19, 31-33]; few of them were adapted for frozen
samples [34]. While these protocols have been well-es-
tablished for the respective sample types, with known
details and critical points [35-37], their applicability
to FFPE tumor sections is limited. Currently, there are
only two Hi-C protocols for the analysis of FFPE sec-
tions [28, 29]. We compared these two existing proto-
cols (Table1) and identified significant methodological
differences, particularly, at the deparaffinization and
sample lysis stages. For deparaffinization, Troll etal. [28]
recommended xylene treatment followed by alcohol
washing, while Allahyar et al. [29] suggested a 3-min
incubation at 80°C, centrifugation, and removal of
the paraffin layer. After deparaffinization, in order
to ensure the availability of chromatin for restriction
enzymes, Troll et al. treated the samples with protein-
ase K (0.5 mg/ml for 1 h at 37°C), whereas Allahyar etal.
used sonication and subsequent incubation for 2 h
at 80°C. Both proteinase treatment and prolonged in-
cubation at 80°C can lead to the destruction of cross-
links formed by formaldehyde [38] and DNA release
from the chromatin. Finally, both studies suggested us-
ing restriction endonucleases with four-nucleotide rec-
ognition sites for chromatin fragmentation. However,
this approach can result in a low coverage of genom-
ic regions distant from the enzyme recognition sites,
which might limit identification of certain genomic
variants, such as SNVs in the oncogene exons located
far from the restriction sites.
We have developed a modified Hi-C protocol spe-
cifically designed for preparing libraries from FFPE
sections (Table 1). The lysis conditions were adjusted
to be less harsh, and we used S1 nuclease instead of
restriction endonucleases (see Materials and methods)
to provide uniform genome coverage [30].
During the preparation of the Hi-C libraries from
living cells or tissues, the quality of DNA fragments is
tested after the following key steps:
1. Post-lysis, pre-fragmentation;
2. Post-fragmentation, pre-ligation;
3. Post-ligation.
These control checkpoints are crucial for eval-
uating the quality of prepared libraries (Fig. 1, a, b).
Before fragmentation, a band corresponding to the
high-molecular-weight DNA should be detected, which
disappears after fragmentation with the formation
of many low-molecular fragments of various lengths.
After ligation, the distribution of fragment lengths
shifts to a higher molecular weight region.
We found that the standard quality controls typ-
ically employed in the Hi-C library preparation from
living tissues were not applicable or representative
Table 1. Comparison of FFPE Hi-C protocols
Steps
Conditions
Troll et al. [28] Allahyar et al. [29] Our protocol
Deparaffinization xylene
3 min at 80°C,
centrifugation
3 min at 80°C,
centrifugation
Lysis proteinase K, 1h at 37°C
0.6% SDS, sonication,
incubation at 80°C for 2 h
lysis in the presence of ionic
and nonionic detergents
Chromatin fragmentation MboI, 1h at 37°C NlaIII, 1 h at 37°C S1 nuclease, 1h at 37°C
Biotin labeling + +
Ligation 2 h, room temperature 1 h, 16°C overnight, 16°C
Ligation product enrichment + +
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 641
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Fig. 1. Results of chromatin digestion and ligation in Hi-C experiments in living cells and tissues using DpnII(a) and S1 nucle-
ase(b) and in FFPE sections using our protocol(c). Lanes: 1,pre-fragmentation, 2,post fragmentation, 3,post-ligation; M,100-bp
ladder (SibEnzyme). d)DNA quantification(ng) in Hi-C libraries obtained from cells and FFPE sections of different tumor types.
The number of analyzed libraries from peripheral blood mononuclear cells (PBMCs) was 16, from FFPE sections: LCL – 13,
CLL– 9, gliomas– 4, sarcomas– 5.
inthe case of FFPE samples. Long-term fixation of tu-
mor tissues in formaldehyde and subsequent embed-
ding in a paraffin block lead to a significant DNA deg-
radation [39, 40]. Consequently, DNA extracted from
FFPE blocks is already in a highly fragmented state.
According to our data, further nuclease treatment and
ligation do not result in contrasting changes in the
fragment length (Fig.1c). However, our analysis of the
sequencing data quality and visual examination of the
resulting FFPE Hi-C maps indicated successful com-
pletion of the key Hi-C protocol stages. For instance,
the Hi-C libraries represented in Fig.1, as well as ad-
ditional libraries detailed in Online Resource 2 (sam-
ples s11-s15), demonstrated acceptable quality metrics.
Hence, we believe that in the case of FFPE Hi-C, the de-
scribed controls are not necessary. Instead, we recom-
mend assessing the library quality based on the results
of shallow sequencing.
Unlike the FFPE Hi-C method, Hi-C analysis of live
cells using restriction endonuclease DpnII or S1 nu-
clease allows to easily vary the amount of the starting
material, while in the case of FFPE sections, accurate
estimation of the number of cells in each section can
be a challenge. We observed a significant variability in
the amount of DNA isolated from FFPE samples of dif-
ferent tumors, as well as in the samples of the same tu-
mor type (Fig.1d). Thus, FFPE sections from sarcomas
consistently yielded the lowest amount of DNA, which
forced us to utilize three FFPE sections from a single
block for analysis. Therefore, we recommend to deter-
mine the required number of sections for each tumor
type to obtain sufficient yield of DNA libraries.
GRIDINA et al.642
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Fig. 2. Quality metrics of FFPE Hi-C datasets showing proportion of unmapped reads(a), proportion of PCR duplicates(b), pro-
portion of DEs(c), proportion of cis contacts among all Hi-C contacts(d). Each dot represents an independent Hi-C library prepa-
ration. The number of analyzed libraries from PBMCs was 16 and from FFPE sections: LCL– 13, CLL– 9, gliomas– 4, sarcomas– 2.
Using the newly developed protocol, we prepared
FFPE Hi-C libraries from CLL, LCL, gliomas, and sar-
coma samples. The CLL sections were also used to
construct the libraries according to the protocols sug-
gested by Troll etal. [28] and Allahyar etal. [29]. After
sequencing, the libraries were assessed for their quali-
ty. We observed a low number of unmapped reads and
PCR duplicates across all the libraries (Fig.2, a and b,
respectively). Another key quality metric for Hi-C li-
braries is a proportion of dangling ends (DEs), unin-
formative fragments that are not ligation products.
The DE content in the Hi-C libraries prepared from
peripheral blood mononuclear cells (PBMCs) using S1
nuclease averaged 40%. However, in the FFPE Hi-C li-
braries prepared according to the developed protocol,
this proportion was higher and varied depending on
the tumor type. The highest DE content was observed
in the libraries prepared following the protocols of
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 643
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Fig. 3. Representative Hi-C data obtained for FFPE sections using the developed protocol. a)Chromosome-scale Hi-C heatmap for
sample s26 (above the diagonal) and the map obtained for blood lymphocytes (control; below the diagonal) [30]. b)Examples
of contact patterns in Hi-C maps and their schematic representation: reciprocal translocations(b), inversions(c), heterozygous
deletions(d), heterozygous deletions and duplications(e). In panels(b-e), above the diagonal, contact map for sample s25; below
the diagonal, contact map for s26 (control).
Troll et al. [28] and Allahyar et al. (Fig. 2c). We have
previously shown [31] that the most important metric
of the library quality is a proportion of cis contacts [cis/
all ratio (FF and RR orient)], which reflects the propor-
tion of Hi-C reads aligned to the same chromosome.
Thus, a low proportion of cis contacts may result from
the reversal of cross-links and subsequent random li-
gation of released DNA. For all the libraries prepared
using our protocol, the proportion of cis contacts was
above 70%, while for the libraries prepared according
to Troll et al. and Allahyar et al., it was 20 and 15%,
respectively (Fig.2d).
However, it was not the aim of our study to com-
pare between our FFPE Hi-C protocol and protocols
proposed by Troll et al. [28] and Allahyar et al. [29],
as the latter ones utilize enzymes with specific recog-
nition sites, which limits their capacity to identify ge-
nomic variants with the efficacy that can be achieved
with our protocol, which uses S1 nuclease [31, 41].
Therefore, we have not used the protocols by Troll
et al. [28] and Allahyar et al. [29] for a larger set of
samples or different tumor types. Although the ob-
tained data did not allow us to draw a statistical com-
parison of the quality of Hi-C data obtained by differ-
ent protocols, we can still state a higher quality of Hi-C
libraries obtained using our protocol.
Two libraries were subjected to deep sequencing
and the results obtained were used to construct heat-
maps of the Hi-C contacts, which revealed distinct
patterns indicating various types of chromosomal re-
arrangements (Fig. 3, a-e). We were able to detect all
types of chromosomal structural rearrangements, such
as deletions, duplications, inversions, and transloca-
tions. Based on these findings, we can conclude that
the developed method for FFPE Hi-C analysis using S1
nuclease was effective in generating maps of three-
GRIDINA et al.644
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
dimensional chromatin contacts and identifying chro-
mosomal rearrangements based on the obtained data.
Prospects of FFPE Hi-C analysis in the selection
of targeted therapy in oncological diseases. The de-
veloped FFPE Hi-C protocol has a significant potential
for future clinical applications, as it can detect chromo-
somal rearrangements (translocations and inversions),
including balanced ones. These rearrangements are
often challenging to identify using standard sequenc-
ing methods. The practical value of molecular genetic
testing lies in its ability to influence patient treatment
strategies and provide prognostic insights. Detection of
specific chromosomal rearrangements can guide the
choice of targeted therapy, which is a critical aspect
of personalized medicine and oncology. To determine
the scope of applications for our method, we analyzed
the databases of the National Cancer Institute (https://
www.cancer.gov/about-cancer/treatment/drugs/
cancer-type), CIViC (https://civicdb.org/welcome), and
MyCancerGenome (https://www.mycancergenome.org/).
We identified balanced chromosomal rearrangements
leading to the gene fusions resulting in the generation
of chimeric products that have been already targeted
in existing or developed therapies (Table2). Our analy-
sis revealed that the highest number of gene fusions
for which the targeted therapy had been developed has
been described for hematological oncological diseases
and, in the case of solid tumors, for various types of
sarcomas and lung cancer. These diseases also exhibit
the greatest diversity of fusion partners. Thus, several
fusion variants have been identified in melanoma and
cholangiocarcinoma; however, in each of the fusions,
one of the partners remained the same: BRAF in mela-
noma and FGFR2 in cholangiocarcinoma.
In addition to the translocations listed in Table2,
there are other rearrangements that are important for
establishing a diagnosis and/or prognosis, but do not
currently have approved targeted drugs. For instance,
KMT2A/MLL gene fusions frequently observed in pa-
tients with acute leukemia, are known to be associ-
ated with a poor prognosis [42]. The list of clinically
relevant gene fusions will expand with the discovery
of new gene fusions and approval of novel therapeutic
drugs.
In order to assess the possibility of detecting chro-
mosomal rearrangements described in Table 2, we
simulated the results of the FFPE Hi-C experiment us-
ing the Charm tool (https://github.com/NuriddinovMA/
Charm). The simulated Hi-C experiments focused on 5
inversions and 7 translocations from Table 2 (Online
Resource 1). The models corresponded to the FFPE
Hi-C libraries sequenced with a depth of about 30
million paired-end reads. For each translocation, we
generated two models: one showed a reciprocal trans-
location and the other showed a translocation of the
minimal-size fragment sufficient to form a chimeric
protein, i.e., a translocation of a region from the 3′-end
of the fused gene to the breakpoint (Fig.4a). The size
of the simulated inversions ranged from 2.3 to 79.9
million bp, while the translocations varied in size from
7.9 to 109 kb. The Hi-C contact heatmaps of clearly
displayed all simulated chromosomal rearrangements
(Fig. 4,b-e; Online Resource 3). This result suggests that
the Hi-C analysis of FFPE samples has a high potential
in detecting known clinically significant chromosomal
rearrangements.
DISCUSSION
In this work, we proposed a protocol for the Hi-C
analysis of FFPE tumor sections using S1 nuclease and
demonstrated that this method can be applied for de-
tecting various types of chromosomal rearrangements.
To fully assess the power of the method, it is crucial
to compare it against other high-throughput diagnos-
tic techniques in terms of several key factors, such as
time, complexity, cost, and accuracy of analysis. Our
protocol can be implemented using standard laborato-
ry equipment commonly used for the preparation of
whole-genome libraries. The process of sample prepa-
ration took 2 more days compared to WGS; however,
the main time-consuming procedure was developing
the logistics for sample pooling and sequencing rather
than sample preparation itself. According to our esti-
mates, the cost of FFPE Hi-C exceeds the cost of WGS
by no more than 20%. A future direction for our re-
search will be understanding the characteristics and
limitations of the FFPE Hi-C method. We still have to
evaluate the sensitivity and specificity of this method,
its ability to determine the proportion of tumor cells in
a sample, and the amount of input material required
for different types of tumors. Then, it will be possible
to fully compare the proposed FFPE Hi-C protocol with
the existing high-throughput diagnostic methods.
The high-throughput sequencing technology has
opened a new era of personalized therapy for oncolo-
gy patients. It has dramatically enhanced our ability to
understand various oncological diseases at a molecular
level. However, it still remains unclear which sequenc-
ing methods and when should be used in daily clinical
practice. In 2020, the European Society of Medical On-
cology recommended routine use of high-throughput
sequencing methods for the diagnostics of non-squa-
mous non-small cell lung cancer, prostate adenocar-
cinoma, ovarian carcinoma, and cholangiocarcinoma
[43]. Despite this limited list, the use of high-through-
put sequencing methods may be worthwhile for other
types of cancer, as it can clarify the diagnosis and even
change the treatment regimen.
A compelling example of the practical applica-
tion of NGS methods for detection of chromosomal
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 645
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Fig. 4. Simulation of chromosomal rearrangements. a)Schematic representation of two variants of translocation leading to
the gene fusion. bandc)Simulated FFPE Hi-C heatmaps for translocations. d-f)Simulated FFPE Hi-C heatmaps for inversions.
Above the diagonal, simulated heatmap; below diagonal, control (simulated FFPE Hi-C map with no structural variations and
with the same number of contacts as the simulated map with the rearrangement).
GRIDINA et al.646
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Table 2. List of balanced chromosomal rearrangements and gene fusions with developed targeted therapy
Rearrangement
Fusion partners
Targeted therapy drugs Diagnosis
AB
t(9;22)(q34;q11)
ABL1
BCR
Dasatinib, Imatinib,
Nilotinib, Bosutinib,
Bafetinib, Asciminib
acute lymphocytic leukemia, acute
myeloid leukemia, chronic myelogenous
leukemia
t(9;12)(q34;p13) ETV6 Imatinib, Dasatinib, Nilotinib B-lymphoblastic leukemia/lymphoma
t(3;9)(p13;q34) FOXP1 Dasatinib B-lymphoblastic leukemia/lymphoma
t(2;9)(q12;q34) RANBP2 Dasatinib B-lymphoblastic leukemia/lymphoma
t(1;9)(q24;q34) RCSD1
Dasatinib, Imatinib,
Ponatinib
B-lymphoblastic leukemia/lymphoma
t(1;9)(p34;q34) SFPQ Dasatinib, Imatinib
acute lymphocytic leukemia, B-lympho-
blastic leukemia/lymphoma
t(5;9)(q23;q34) SNX2 Dasatinib, Imatinib B-lymphoblastic leukemia/lymphoma
inv(1)(q24q25)
ABL2
RCSD1 Imatinib B-lymphoblastic leukemia/lymphoma
t(1;7)(q25;q34) ZC3HAV1 Imatinib B-lymphoblastic leukemia/lymphoma
ALK
different
ALK signaling pathway
inhibitors, HSP90 inhibitors,
EGFR tyrosine kinase
inhibitors, Crizotinib,
Brigatinib, Alectinib,
Ceritinib, Entrectinib,
Pemetrexed, 17-AAG*
breast cancer, colorectal adenocarcino-
ma, epithelioid inflammation, myofibro-
blastic sarcoma, inflammatory myofi-
broblastic tumor, lung adenocarcinoma,
non-small cell lung cancer, thyroid
carcinoma, vaginal sarcoma, anaplastic
large cell lymphoma, diffuse large B-cell
lymphoma
inv(2)(p23;p23) CAD Entrectinib colorectal cancer
t(2;17)(p23;q23) CLTC Crizotinib diffuse large B-cell lymphoma
inv(2)(p23p21) EML4
17-AAG*, Alectinib
(CH5424802), AUY922*,
Ceritinib, Crizotinib,
Brigatinib, IPI-504*,
Erlotinib, Lorlatinib
acinar adenocarcinoma of the lung,
non-small cell lung cancer, renal cell
carcinoma
t(2;7)(p23;q11) HIP1 Crizotinib, Alectinib non-small cell lung cancer
t(2;5)(p23;q35) NPM Crizotinib anaplastic large cell lymphoma
inv(2)(p23q13) RANBP2 Crizotinib inflammatory myofibroblastic tumor
inv(2)(q21;q22) ASNS KMT2E Asparaginase T-cell lymphoblastic leukemia
t(X;17)(p11;q25) TFE3 ASPL Cabozantinib, Dasatinib alveolar soft tissue sarcoma
BRAF
different Cobimetinib, Trametinib melanoma, ovarian cancer
inv(7)(q34;q34) AGK Sorafenib, Vemurafenib melanoma
inv(7)(q21q34) AKAP9 MEK inhibitors papillary thyroid cancer
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 647
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Table 2 (cont.)
Rearrangement
Fusion partners
Targeted therapy drugs Diagnosis
AB
inv(7)(q34;q36)
BRAF
CUL1 MEK inhibitors serous ovarian carcinoma
inv(7)(q22;q34) CUX1 Vemurafenib pancreas cancer
t(1;7)(p31;q34) MIGA1 BRAF Inhibitor, HL-085 Langerhans cell histiocytosis
inv(7)(q32;q34) NRF1 Trametinib transitional cell carcinoma
inv(7)(q33;q34) TRIM24 Vemurafenib melanoma
inv(7)(q22;q34) ZKSCAN1 Trametinib melanoma
t(7;11)(q34;p15) PPFIBP2 Trametinib melanoma
t(4;7)(q25;q34) PAPSS1 Trametinib, Vemurafenib melanoma
t(15;19)(q14;p13) BRD4 NUTM1 Birabresib NUT midline carcinoma
t(X;14)(p22;q32) CRLF2 IGH Ruxolitinib
pediatric B-cell acute lymphoblastic leu-
kemia, adult B-cell acute lymphoblastic
leukemia
t(1;5)(q22;q32) CSF1R MEF2D Imatinib acute lymphocytic leukemia
t(X;6)(q28;p22) DEK AFF2 Pembrolizumab head and neck cancer
t(7;15)(p11;q15) EGFR RAD51
Erlotinib, Osimertinib, Ico-
tinib, Afatinib
lung adenocarcinoma
EWSR1
different TK216* soft tissue sarcoma
t(12;22)(q13;12) ATF1 Crizotinib clear cell sarcoma
t(11;22)(q24;q12) FLI1 genotoxic chemotherapies Ewing sarcoma
t(9;22)(q22;q12) NR4A3 Pazopanib, Sunitinib extraskeletal myxoid chondrosarcoma
inv(8)(p11;p11)
FGFR1
BAG4 AZD4547* carcinoma
t(8;12)(p11;p11) FGFR1OP2 Ponatinib acute myeloid leukemia
t(8;13)(p11;q11) ZNF198 Midostaurin chronic myeloproliferative disease
FGFR2
different
BGJ-398 (Infigratinib),
AZD4575*, JNJ-42756493
(Balversa), Debio1347*, Futi-
batinib, Erdafitinib, Infigrati-
nib, Phosphate, Pemigatinib
cholangiocarcinoma, bladder cancer
t(1;10)(p13;q26) AHCYL1 BGJ398 cholangiocarcinoma
inv(10)(q21;q26) BICC1 BGJ398, JNJ-42756493
cholangiocarcinoma, endometrial can-
cer, urothelial carcinoma
t(4;10)(p16;q26) TACC3 Pazopanib, Ponatinib cholangiocarcinoma
GRIDINA et al.648
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Table 2 (cont.)
Rearrangement
Fusion partners
Targeted therapy drugs Diagnosis
AB
FGFR3 different
FGFR signaling blockers,
TORC1/2 inhibitors, AZD4547*,
JNJ-42756493, BGJ398,
Debio1347*, Erdafitinib
bladder cancer
FLT3 different
FLT3 signaling blockers,
Sorafenib, all-trans
retinoic acid, Anthracycline*,
Daunorubicin*, Lestaurtinib,
Selumetinib (AZD6244)
acute myeloid leukemia
t(2;13)(q36;q14) FOXO1 PAX3 Thapsigargin alveolar rhabdomyosarcoma
t(9;22)(p24;q11)
JAK2
BCR Ruxolitinib myeloid neoplasm
t(9;14)(p24;q32) GOLGA5 Ruxolitinib b-lymphoblastic leukemia/lymphoma
t(8;9)(p22;p24) PCM1 Ruxolitinib leukemia
t(5;9)(q14;p24) SSBP2 Ruxolitinib B-lymphoblastic leukemia/lymphoma
KIT different Imatinib malignant melanoma of the rectum
inv(7)(q22;q31)
MET
ATXN7L1 Crizotinib non-small cell lung cancer
t(7;10)(q31;p11) KIF5B Crizotinib Non-small cell lung cancer
t(7;8)(q31;p12) RBPMS Cabozantinib congenital fibrosarcoma
t(8;21)(q24,p12)
NRG1
APP Afatinib
t(1;8)(q24;p12) ATP1B1 Afatinib
cholangiocarcinoma, pancreatic ductal
adenocarcinoma
t(5;8)(q33;p12) CD74 Afatinib mucinous adenocarcinoma
t(8;11)(p12;q12) SLC3A2
Afatinib, Erlotinib,
Lumretuzumab
lung adenocarcinoma, mucinous adeno-
carcinoma
t(8;20)(p12q13) SDC4 Afatinib lung adenocarcinoma
NTRK1
different
ARRY-470, Entrectinib,
Larotrectinib (LOXO-101),
LESTAURTINIB*, Crizotinib
colorectal cancer, sarcoma, all solid
tumors, lung adenocarcinoma
t(1;12)(q23;p13) ETV6 Entrectinib all types
t(1;5)(q23;q35) SQSTM1 Entrectinib non-small cell lung cancer
inv(1)(q21q23) TPM3 Larotrectinib, Entrectinib Sarcoma, papillary thyroid cancer
inv(1)(q23q31) TPR Larotrectinib all solid tumors
NTRK2 different Larotrectinib, Entrectinib all solid tumors
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 649
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
Table 2 (cont.)
Rearrangement
Fusion partners
Targeted therapy drugs Diagnosis
AB
NTRK3
different
Entrectinib, Larotrectinib,
blockers of PI3K, SRC, and
IGF1 signaling pathways
all solid tumors
t(12;15)(p13;q25) ETV6
Entrectinib, Etoposide,
Larotrectinib, Crizotinib
salivary gland carcinoma, breast cancer,
congenital mesoblastic nephroma,
B-lymphoblastic leukemia/lymphoma
PDGFRA
different Imatinib
myelodysplasia, myeloproliferative
neoplasms
t(4;22)(q12;q11) BCR Imatinib B-cell acute lymphoblastic leukemia
PDGFRB
different Imatinib
chronic myeloproliferative disease,
dermatofibrosarcoma, myelodysplasia,
myeloproliferative neoplasm, acute lym-
phocytic leukemia
t(12;17)(p13;q21) ATF7IP
Dasatinib, Imatinib, Nilo-
tinib, Ponatinib, Bafetinib,
Rebastinib
pediatric B-cell acute lymphoblastic
leukemia
t(17;22)(q21;q13) COL1A1 Sunitinib dermatofibrosarcoma
t(15;17)(q22;q21) PML RARA
Tretinoin, arsenic trioxide,
all-trans-retinoic acid
acute promyelocytic leukemia
RET
different
Cabozantinib, Vandetanib,
Pralsetinib, Selpercatinib
non-small cell lung cancer, thyroid
cancer
inv(10)(q11q21) CCDC6
Nintedanib, Agerafenib,
Pralsetinib
non-small cell lung cancer, colorectal
cancer, solid tumors
inv(10)(p11q11) KIF5B
Vandetanib, Selpercatinib,
Everolimus, Pralsetinib
lung adenocarcinoma, solid tumors
ROS1
different
Ceritinib, Entrectinib, Erlo-
tinib, Gefitinib
lung adenocarcinoma, bronchioloalve-
olar adenocarcinoma, colorectal adeno-
carcinoma, non-small cell lung cancer
t(5;6)(q33;q22) CD74
Cabozantinib, Crizotinib, Bri-
gatinib, Ceritinib, Foretinib,
Lorlatinib,
non-small cell lung carcinoma, lung
adenocarcinoma
t(3;6)(q12;q22) TFG Crizotinib inflammatory myofibroblastic tumor
RSPO2 different
Wnt signaling inhibitors,
CGX1321
digestive system cancer
Note. We call the fusion partner “A” the gene, the changes in the activity of which lead to the disease development.
* At the clinical trial stage.
rearrangements in clinical oncology was reported by
Hehir-Kwa etal. [44]. Based on histological analysis of
brain tumor, a one-year-old child was diagnosed with
glioblastoma. In accordance with the diagnosis, the
treatment was started using the HGG HIT protocol for
infants. After 6 weeks of active tumor growth, the pro-
tocol was changed to BBSFOP HGG. DNA methylation
analysis allowed the tumor to be classified as a glioma,
GRIDINA et al.650
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
and RNA sequencing revealed a fusion of the ZCCHC8
and ROS1 genes resulting from the translocation of
chromosomes 6 and 12. After fifteen months of treat-
ment, the tumor continued to grow and metastases
appeared. After consultation and obtaining a consent
from the child’s parents, treatment with the ROS1 ty-
rosine kinase inhibitor Entrectinib was initiated. Re-
markably, within a month from starting this targeted
therapy, the tumor size significantly decreased [44].
While this case represents a notable success, it
remains an exception in the current landscape of tar-
geted cancer therapy. Presently, targeted therapies are
predominantly developed for diseases characterized
by point mutations or gene fusions resulting in chi-
meric proteins. This is mostly due to the limitations
of existing detection methods, which are less adept at
identifying balanced chromosomal rearrangements,
particularly those occurring in non-coding regions.
Analysis of FFPE samples using Hi-C is capable of de-
tecting not only chromosomal rearrangements leading
to gene fusions, but also those that occur in non-cod-
ing regions and can disrupt regulatory element, which
might alter oncogene expression [45]. The application
of the Hi-C technique developed by us offers an exten-
sion of already existing high-throughput sequencing
methods. By enabling detection of a broader range of
genomic alterations, it allows researchers to uncover
clinically relevant information that might otherwise
remain elusive. This comprehensive genomic insight is
instrumental not only in promoting our understanding
of tumor biology, but also in identifying novel thera-
peutic targets.
Supplementary information. The online version
contains supplementary material available at https://
doi.org/10.1134/S0006297924040047.
Acknowledgments. We express our gratitude to
the Ministry of Science and Higher Education of the
Russian Federation (state project FWNR-2022-0019)
for providing access to computing power. Access to the
public resources and datasets was provided by Novo-
sibirsk State University with the support of the Minis
Ministry of Science and Higher Education of the Rus-
sian Federation (grant no.2019-0546; FSUS-2024-0018).
Contributions. M.M.G. and V.S.F. developed the
concept and managed the study; M.M.G., Ya.K.S., M.A.N.,
T.A.L., N.Yu.T., A.A.S., A.I.R., N.V.V. , S.V.V., T.S.G., E.V.D.,
and M.A.T. conducted experiments; M.M.G., V.S.F., E.V.D.,
and M.A.K. discussed the results; M.M.G. and Ya.K.S.
wrote the manuscript; V.S.F. edited the text of the
article.
Funding. The research was supported by the Rus-
sian Science Foundation (project no.22-24-00190).
Ethics declarations. This study was performed in
line with the principles of the Declaration of Helsinki.
Approval was granted by the Research Ethics Com-
mittee of the Research Institute of Medical Genetics,
Tomsk National Research Medical Center (27.07.2017/
no. 106). All study participants provided informed con-
sent. The authors of this work declare that they have
no conflicts of interest.
REFERENCES
1. Schmitz, R., Ceribelli, M., Pittaluga, S., Wright, G.,
and Staudt, L. M. (2014) Oncogenic mechanisms in
Burkitt lymphoma, Cold Spring Harb. Perspect. Med.,
4, a014282, doi:10.1101/cshperspect.a014282.
2. Kumar-Sinha,C., Tomlins, S.A., and Chinnaiyan, A.M.
(2008) Recurrent gene fusions in prostate cancer, Nat.
Rev. Cancer, 8, 497-511, doi:10.1038/nrc2402.
3. Pflueger, D., Rickman, D. S., Sboner, A., Perner, S.,
LaFargue, C. J., Svensson, M. A., Moss, B. J., Kitaba-
yashi, N., Pan, Y., de la Taille, A., Kuefer, R., Tewari,
A. K., Demichelis, F., Chee, M. S., Gerstein, M. B., and
Rubin, M. A. (2009) N-Myc downstream regulated
gene 1 (NDRG1) is fused to ERG in prostate cancer,
Neoplasia, 11, 804-811, doi:10.1593/neo.09572.
4. Sorensen, P.H., Lessnick, S.L., Lopez-Terrada,D., Liu,
X.F., Triche, T.J., and Denny, C.T. (1994) A second Ew-
ing’s sarcoma translocation, t(21,22), fuses the EWS
gene to another ETS-family transcription factor, ERG,
Nat. Genet., 6, 146-151, doi:10.1038/ng0294-146.
5. Sotoca, A.M., Prange, K.H. M., Reijnders, B., Mando-
li,A., Nguyen, L.N., Stunnenberg, H.G., and Martens,
J. H. A. (2016) The oncofusion protein FUS-ERG tar-
gets key hematopoietic regulators and modulates the
all-trans retinoic acid signaling pathway in t(16,21)
acute myeloid leukemia, Oncogene, 35, 1965-1976,
doi:10.1038/onc.2015.261.
6. Persson, M., Andrén, Y., Mark, J., Horlings, H. M.,
Persson, F., and Stenman, G. (2009) Recurrent fu-
sion of MYB and NFIB transcription factor genes in
carcinomas of the breast and head and neck, Proc.
Natl. Acad. Sci. USA, 106, 18740-18744, doi: 10.1073/
pnas.0909114106.
7. Humtsoe, J. O., Kim, H.-S., Jones, L., Cevallos, J., Boi-
leau,P., Kuo,F., Morris, L.G.T., and Ha,P. (2022) De-
velopment and characterization of MYB-NFIB fusion
expression in adenoid cystic carcinoma, Cancers, 14,
2263, doi:10.3390/cancers14092263.
8. Turner,N., and Grose,R. (2010) Fibroblast growth fac-
tor signalling: from development to cancer, Nat. Rev.
Cancer, 10, 116-129, doi:10.1038/nrc2780.
9. Nakanishi, Y., Akiyama, N., Tsukaguchi, T., Fujii, T.,
Satoh,Y., Ishii,N., and Aoki,M. (2015) Mechanism of
oncogenic signal activation by the novel fusion ki-
nase FGFR3-BAIAP2L1, Mol. Cancer Ther., 14, 704-712,
doi:10.1158/1535-7163.MCT-14-0927-T.
10. Guo,Q., Lakatos, E., Bakir, I. A., Curtius, K., Graham,
T. A., and Mustonen, V. (2022) The mutational signa-
HI-C ANALYSIS OF FFPE TUMOR SECTIONS 651
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
tures of formalin fixation on the human genome, Nat.
Commun., 13, 4487, doi:10.1038/s41467-022-32041-5.
11. Scolnick, J. A., Dimon, M., Wang, I.-C., Huelga, S. C.,
and Amorese, D. A. (2015) An efficient method for
identifying gene fusions by targeted RNA sequencing
from fresh frozen and FFPE samples, PLoS One, 10,
e0128916, doi:10.1371/journal.pone.0128916.
12. Mardis, E.R., and Wilson, R.K. (2009) Cancer genome
sequencing: a review, Hum. Mol. Genet., 18, 163-168,
doi:10.1093/hmg/ddp396.
13. Maher, C.A., Kumar-Sinha,C., Cao,X., Kalyana-Sund-
aram,S., Han,B., Jing,X., Sam,L., Barrette,T., Palani-
samy,N., and Chinnaiyan, A.M. (2009) Transcriptome
sequencing to detect gene fusions in cancer, Nature,
458, 97-101, doi:10.1038/nature07638.
14. Peng,H., Huang,R., Wang,K., Wang,C., Li,B., Guo,Y.,
Li,M., Zhang,D., Dong,H., Chen,H., Chen, C., Xu, Q.,
Li, F., Tian, L., and Wu, J. (2021) Development and
validation of an RNA sequencing assay for gene fu-
sion detection in formalin-fixed, paraffin-embed-
ded tumors, J. Mol. Diagn., 23, 223-233, doi: 10.1016/
j.jmoldx.2020.11.005.
15. Ahlfen, S. vo., Missel, A., Bendrat, K., and Schlump-
berger, M. (2007) Determinants of RNA quality from
FFPE samples, PLoS One, 2, e1261, doi:10.1371/journal.
pone.0001261.
16. Groelz, D., Sobin, L., Branton, P., Compton, C.,
Wyrich, R., and Rainen, L. (2013) Non-formalin fixa-
tive versus formalin-fixed tissue: a comparison of his-
tology and RNA quality, Exp. Mol. Pathol., 94, 188-194,
doi:10.1016/j.yexmp.2012.07.002.
17. Wang,X., Xu,J., Zhang,B., Hou,Y., Song,F., Lyu,H., and
Yue, F. (2021) Genome-wide detection of enhancer-
hijacking events from chromatin interaction data
in re-arranged genomes, Nat. Methods, 18, 661-668,
doi:10.1038/s41592-021-01164-w.
18. Engreitz, J. M., Agarwala, V., and Mirny, L. A. (2012)
Three-dimensional genome architecture influences
partner selection for chromosomal translocations
in human disease, PLoS One, 7, e44196, doi:10.1371/
journal.pone.0044196.
19. Harewood,L., Kishore,K., Eldridge, M.D., Wingett,S.,
Pearson, D., Schoenfelder, S., Collins, V. P., and Fra-
ser, P. (2017) Hi-C as a tool for precise detection and
characterisation of chromosomal rearrangements and
copy number variation in human tumours, Genome
Biol., 18, 125, doi:10.1186/s13059-017-1253-8.
20. Chakraborty, A., and Ay, F. (2017) Identification of
copy number variations and translocations in can-
cer cells from Hi-C data, Bioinformatics, 34, 338-345,
doi:10.1093/bioinformatics/btx664.
21. Dixon, J.R., Xu,J., Dileep,V., Zhan,Y., Song,F., Le, V.T.,
Yardımcı, G.G., Chakraborty,A., Bann, D.V., Wang,Y.,
Clark,R., Zhang,L., Yang,H., Liu,T., Iyyanki,S., An,L.,
Pool, C., Sasaki, T., Rivera-Mulia, J. C., Ozadam, H.,
Lajoie, B.R., Kaul,R., Buckley,M., Lee,K., Diegel,M.,
Pezic, D., Ernst, C., Hadjur, S., Odom, D.T., Stamatoy-
annopoulos, J.A., Broach, J.R., Hardison, R.C., Ay,F.,
Noble, W.S., Dekker,J., Gilbert, D.M., and Yue,F. (2018)
Integrative detection and analysis of structural vari-
ation in cancer genomes, Nat. Genet., 50, 1388-1398,
doi:10.1038/s41588-018-0195-8.
22. Melo, U. S., Schöpflin, R., Acuna-Hidalgo,R., Mensah,
M.A., Fischer-Zirnsak,B., Holtgrewe,M., Klever, M.-K.,
Türkmen, S., Heinrich, V., Pluym, I. D., Matoso, E.,
Bernardo de Sousa, S., Louro, P., Hülsemann, W., Co-
hen, M., Dufke, A., Latos-Bieleńska, A., Vingron, M.,
Kalscheuer, V., Quintero-Rivera, F., Spielmann, M.,
and Mundlos, S. (2020) Hi-C identifies complex ge-
nomic rearrangements and TAD-shuffling in devel-
opmental diseases, Am. J. Hum. Genet.
, 106, 872-884,
doi:10.1016/j.ajhg.2020.04.016.
23. Adeel, M. M., Rehman, K., Zhang, Y., Arega, Y., and
Li, G. (2022) Chromosomal translocations detection
in cancer cells using chromosomal conformation cap-
ture data, Genes, 13, 1170, doi:10.3390/genes13071170.
24. Du,Y., Gu,Z., Li,Z., Yuan,Z., Zhao,Y., Zheng,X., Bo,X.,
Chen, H., and Wang, C. (2022) Dynamic interplay be-
tween structural variations and 3D genome orga-
nization in pancreatic cancer, Adv. Sci., 9, 2200818,
doi:10.1002/advs.202200818.
25. Kim,K., Kim, M., Kim, Y., Lee, D., and Jung, I. (2022)
Hi-C as a molecular rangefinder to examine genomic
rearrangements, Semin. Cell Dev. Biol., 121, 161-170,
doi:10.1016/j.semcdb.2021.04.024.
26. Song, F., Xu, J., Dixon, J., and Yue, F. (2022) Analysis
of Hi-C data for discovery of structural variations in
cancer, Methods Mol. Biol., 2301, 143-161, doi:10.1007/
978-1-0716-1390-0_7.
27. Sidiropoulos, N., Mardin, B. R., Rodríguez-González,
F.G., Bochkov, I.D., Garg,S., Stütz, A.M., Korbel, J.O.,
Aiden, E. L., and Weischenfeldt, J. (2022) Somatic
structural variant formation is guided by and influ-
ences genome architecture, Genome Res., 32, 643-655,
doi:10.1101/gr.275790.121.
28. Troll, C. J., Putnam, N. H., Hartley, P. D., Rice, B.,
Blanchette, M., Siddiqui, S., Ganbat, J.-O., Powers,
M. P., Ramakrishnan, R., Kunder, C. A., Bustaman-
te, C. D., Zehnder, J. L., Green, R. E., and Costa, H. A.
(2019) Structural variation detection by proximity
ligation from formalin-fixed, paraffin-embedded tu-
mor tissue, J. Mol. Diagn., 21, 375-383, doi: 10.1016/
j.jmoldx.2018.11.003.
29. Allahyar, A., Pieterse, M., Swennenhuis, J., Los-de
Vries, G.T., Yilmaz,M., Leguit,R., Meijers, R.W.J., van
der Geize, R., Vermaat, J., Cleven, A., van Wezel, T.,
Diepstra,A., van Kempen, L.C., Hijmering, N. J., Sta-
thi,P., Sharma,M., Melquiond, A.S.J., de Vree, P.J.P.,
Verstegen, M.J.A.M., Krijger, P.H.L., Hajo,K., Simo-
nis,M., Rakszewska,A., van Min,M., de Jong,D., Yls-
tra,B., Feitsma,H., Splinter,E., and de Laat,W. (2021)
Robust detection of translocations in lymphoma FFPE
GRIDINA et al.652
BIOCHEMISTRY (Moscow) Vol. 89 No. 4 2024
samples using targeted locus capture-based sequenc-
ing, Nat. Commun., 12, 3361, doi: 10.1038/s41467-
021-23695-8.
30. Gridina, M., Popov, A., Shadskiy, A., Torgunakov, N.,
Kechin, A., Khrapov, E., Ryzhkova, O., Filipenko, M.,
and Fishman, V. (2023) Expanding the list of se-
quence-agnostic enzymes for chromatin confor-
mation capture assays with S1 nuclease, bioRxiv,
doi:10.1101/2023.06.15.545138.
31. Gridina,M., Mozheiko,E., Valeev,E., Nazarenko, L.P.,
Lopatkina, M. E., Markova, Z. G., Yablonskaya, M. I.,
Voinova, V. Y., Shilova, N. V., Lebedev, I.N., and Fish-
man, V. (2021) A cookbook for DNase Hi-C, Epigenet.
Chromatin, 14, 15, doi:10.1186/s13072-021-00389-5.
32. Rao, S. S. P., Huntley, M. H., Durand, N. C., Stameno-
va, E.K., Bochkov, I.D., Robinson, J.T., Sanborn, A.L.,
Machol,I., Omer, A. D., Lander, E. S., and Aiden, E. L.
(2014) A 3D map of the human genome at kilobase res-
olution reveals principles of chromatin looping, Cell,
159, 1665-1680, doi:10.1016/j.cell.2014.11.021.
33. Belaghzal, H., Dekker, J., and Gibcus, J. H. (2017)
Hi-C 2.0: An optimized Hi-C procedure for high-res-
olution genome-wide mapping of chromosome con-
formation, Methods, 123, 56-65, doi: 10.1016/j.ymeth.
2017.04.004.
34. Zheng,W., Yang, Z., Ge, X., Feng,Y., Wang, Y., Liu,C.,
Luan,Y., Cai,K., Vakal, S., You, F., Guo,W., Wang,W.,
Feng, Z., and Li, F. (2021) Freeze substitution Hi-C, a
convenient and cost-effective method for capturing
the natural 3D chromatin conformation from frozen
samples, J.Genet. Genomics, 48, 237-247, doi:10.1016/
j.jgg.2020.11.002.
35. Golloshi,R., Sanders, J.T., and McCord, R.P. (2018) Iter-
atively improving Hi-C experiments one step at a time,
Methods, 142, 47-58, doi:10.1016/j.ymeth.2018.04.033.
36. Lafontaine, D.L., Yang,L., Dekker,J., and Gibcus, J.H.
(2021) Hi-C 3.0: improved protocol for genome-wide
chromosome conformation capture, Curr. Protoc.,
1, e198, doi:10.1002/cpz1.198.
37. Akgol Oksuz, B., Yang, L., Abraham, S., Venev, S. V.,
Krietenstein, N., Parsi, K. M., Ozadam, H., Oomen,
M. E., Nand, A., Mao, H., Genga, R. M. J., Maehr, R.,
Rando, O.J., Mirny, L.A., Gibcus, J.H., and Dekker, J.
(2021) Systematic evaluation of chromosome confor-
mation capture assays, Nat. Methods, 18, 1046-1055,
doi:10.1038/s41592-021-01248-7.
38. Kennedy-Darling, J., and Smith, L. M. (2014) Mea-
suring the formaldehyde protein-DNA cross-link re-
versal rate, Anal. Chem., 86, 5678-5681, doi: 10.1021/
ac501354y.
39. Einaga, N., Yoshida, A., Noda, H., Suemitsu, M., Na-
kayama, Y., Sakurada, A., Kawaji, Y., Yamaguchi, H.,
Sasaki,Y., Tokino,T., and Esumi,M. (2017) Assessment
of the quality of DNA from various formalin-fixed par-
affin-embedded (FFPE) tissues and the use of this DNA
for next-generation sequencing (NGS) with no artifac-
tual mutation, PLoS One, 12, e0176280, doi: 10.1371/
journal.pone.0176280.
40. Kuwata, T., Wakabayashi, M., Hatanaka, Y., Morii, E.,
Oda, Y., Taguchi,K., Noguchi, M., Ishikawa, Y., Naka-
jima,T., Sekine,S., Nomura,S., Okamoto,W., Fujii,S.,
and Yoshino,T. (2020) Impact of DNA integrity on the
success rate of tissue-based next-generation sequenc-
ing: lessons from nationwide cancer genome screen-
ing project SCRUM-Japan GI‐SCREEN, Pathol. Int.,
70, 932-942, doi:10.1111/pin.13029.
41. Ma, W., Ay, F., Lee, C., Gulsoy, G., Deng, X., Cook, S.,
Hesson, J., Cavanaugh, C., Ware, C. B., Krumm, A.,
Shendure,J., Blau, C.A., Disteche, C.M., Noble, W.S.,
and Duan, Z. (2018) Using DNase Hi-C techniques to
map global and local three-dimensional genome ar-
chitecture at high resolution, Methods, 142, 59-73,
doi:10.1016/j.ymeth.2018.01.014.
42. Lomov, N., Zerkalenkova, E., Lebedeva, S., Viush-
kov, V., and Rubtsov, M. A. (2021) Cytogenetic and
molecular genetic methods for chromosomal translo-
cations detection with reference to the KMT2A/MLL
gene, Crit. Rev. Clin. Lab. Sci., 58, 180-206, doi:10.1080/
10408363.2020.1844135.
43. Mosele, F., Remon, J., Mateo, J., Westphalen, C. B.,
Barlesi, F., Lolkema, M. P., Normanno, N., Scarpa, A.,
Robson, M., Meric-Bernstam, F., Wagle, N., Stenzing-
er, A., Bonastre, J., Bayle, A., Michiels, S., Bièche, I.,
Rouleau,E., Jezdic,S., Douillard, J.-Y., Reis-Filho, J.S.,
Dienstmann, R., and André, F. (2020) Recommenda-
tions for the use of next-generation sequencing (NGS)
for patients with metastatic cancers: a report from the
ESMO precision medicine working group, Ann. Oncol.,
31, 1491-1505, doi:10.1016/j.annonc.2020.07.014.
44. Hehir-Kwa, J.Y., Koudijs, M.J., Verwiel, E.T.P., Kester,
L. A., van Tuil, M., Strengman, E., Buijs, A., Kranen-
donk, M.E.G., Hiemcke-Jiwa, L.S., de Haas,V., van de
Geer,E., de Leng,W., van der Lugt,J., Lijnzaad,P., Hol-
stege, F. C. P., Kemmeren, P., and Tops, B. B. J. (2022)
Improved gene fusion detection in childhood cancer
diagnostics using RNA sequencing, JCO Precis. Oncol.,
6, e2000504, doi:10.1200/PO.20.00504.
45. Zhang, Y., Chen, F., and Creighton, C. J. (2021) SVEx-
press: identifying gene features altered recurrently
in expression with nearby structural variant break-
points, BMC Bioinformatics, 22, 135, doi: 10.1186/
s12859-021-04072-0.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.