ArticlePDF Available

Suppression of BRAFV599E in Human Melanoma Abrogates Transformation

Authors:

Abstract and Figures

Activating mutations in the BRAF serine/threonine kinase are found in >70% of human melanomas, of which >90% are BRAF(V599E). We sought to investigate the role of the BRAF(V599E) allele in malignant melanoma. We here report that suppression of BRAF(V599E) expression by RNA interference in cultured human melanoma cells inhibits the mitogen-activated protein kinase cascade, causes growth arrest, and promotes apoptosis. Furthermore, knockdown of BRAF(V599E) expression completely abrogates the transformed phenotype as assessed by colony formation in soft agar. Similar targeting of BRAF(V599E) or wild-type BRAF in human fibrosarcoma cells that lack the BRAF(V599E) mutation does not recapitulate these effects. Moreover, these results are specific for BRAF, as targeted interference of CRAF in melanoma cells does not significantly alter their biological properties. Thus, when present, BRAF(V599E) appears to be essential for melanoma cell viability and transformation and, therefore, represents an attractive therapeutic target in the majority of melanomas that harbor the mutation.
Content may be subject to copyright.
[CANCER RESEARCH 63, 5198–5202, September 1, 2003]
Advances in Brief
Suppression of BRAF
V599E
in Human Melanoma Abrogates Transformation
1
Sunil R. Hingorani, Michael A. Jacobetz, Gavin P. Robertson, Meenhard Herlyn, and David A. Tuveson
2
Abramson Family Cancer Research Institute and Abramson Cancer Center at the University of Pennsylvania [S. R. H., M. A. J., D. A. T.] and Department of Medicine [S. R. H.,
D. A. T.], University of Pennsylvania and Wistar Institute [M. H.], Philadelphia, Pennsylvania 19104, and Departments of Pharmacology, Dermatology, and Pathology,
Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 [G. P. R.]
Abstract
Activating mutations in the BRAF serine/threonine kinase are found in
>70% of human melanomas, of which >90% are BRAF
V599E
. We sought
to investigate the role of the BRAF
V599E
allele in malignant melanoma. We
here report that suppression of BRAF
V599E
expression by RNA interfer
-
ence in cultured human melanoma cells inhibits the mitogen-activated
protein kinase cascade, causes growth arrest, and promotes apoptosis.
Furthermore, knockdown of BRAF
V599E
expression completely abrogates
the transformed phenotype as assessed by colony formation in soft agar.
Similar targeting of BRAF
V599E
or wild-type BRAF in human fibrosar
-
coma cells that lack the BRAF
V599E
mutation does not recapitulate these
effects. Moreover, these results are specific for BRAF, as targeted inter-
ference of CRAF in melanoma cells does not significantly alter their
biological properties. Thus, when present, BRAF
V599E
appears to be es
-
sential for melanoma cell viability and transformation and, therefore,
represents an attractive therapeutic target in the majority of melanomas
that harbor the mutation.
Introduction
Malignant melanoma will afflict 50,000 people in the United
States this year and result in 7,000 deaths (1). The incidence of
melanoma is rising among the most rapidly of all malignancies (2).
When diagnosed early, melanoma is highly curable by wide surgical
excision. However, in patients with deep local invasion, or with
spread to lymph nodes or distant sites, the disease is highly resistant
to all forms of therapy. The median survival for patients with meta-
static melanoma is 6–9 months (3).
Recently, activating mutations in the BRAF gene were described in
a majority of melanomas and benign nevi, suggesting an important
role for this oncogene in melanocyte biology and disease (46). More
than 60% of malignant melanomas were found to contain a specific
mutation, BRAF
V599E
, the product of which possesses constitutive
kinase activity. BRAF is a member of the Raf family of serine/
threonine kinases, along with CRAF and ARAF, which serve as
immediate effectors of the ras GTPases (7). Activation of the Raf/
MEK
3
/ERK, or MAPK, signaling cascade promotes cellular prolifer-
ation and survival. The highly homologous Raf family members have
overlapping but distinct biochemical activities and biological func-
tions. We therefore sought to determine whether Raf family members,
and specifically BRAF
V599E
, are required in melanoma cells for main
-
tenance of the transformed state. Accordingly, the biochemical sig-
naling properties and cellular phenotypes of melanoma cells were
assessed after depletion of B-Raf, B-Raf
V599E
, and C-Raf proteins
by RNAi.
Materials and Methods
RNAi Sequences and Preparation. Small inhibitory duplex RNAs (PRO-
LIGO, Boulder, CO) were prepared and reconstituted in annealing buffer as
described (8, 9). The sense strands of the siRNA duplexes were as follows:
Lamin A/C: CUggACUUCCAgAAgAACATT; Com-4: AgAAUUggAUCUg-
gAUCAUTT; Mu-A: gCUACAgAgAAAUCUCgAUTT; C1: UgUgC-
gAAAUggAAUgAgCTT. Duplex shRNA oligos were cloned into the HindIII
and BglII sites in pSUPER.retro (Oligoengine, Seattle, WA), and insert fidelity
was confirmed by sequencing both strands with the following primers: forward
ttatccagccctcactcc; reverse gtgttctgggaaatcacc. The sense strands of the
shRNA pSUPER.retro inserts were as follows:
Com-1: gatccccTGGATACCGTTACATCTTCttcaagagaGAAGATGTAA
CGGTATCCAtttttggaaa.
Com-2: gatccccTCCCAGAGTGCTGTGCTGTttcaagagaACAGCACAG-
CACTCTGGGAtttttggaaa.
Com-3: gatccccTTGGTTGGGACACTGATATttcaagagaATATCAGTG-
TCCCAACCAAtttttggaaa.
Com-4: gatccccAGAATTGGATCTGGATCATttcaagagaATGATCCAG-
ATCCAATTCTtttttggaaa.
Mu-A: gatccccGCTACAGAGAAATCTCGATttcaagagaATCGAGATT-
TCTCTGTAGCtttttggaaa.
Mu-B: gatccccGAGAAATCTCGATGGAGTGttcaagagaCACTCCATC-
GAGATTTCTCtttttggaaa.
C1: gatccccTGTGCGAAATGGAATGAGCttcaagagaGCTCATTCCATT-
TCGCACAtttttggaaa.
BRAF cDNA. Human wild-type BRAF and BRAF
V599E
were cloned from
mRNA and sequenced to confirm fidelity. 5 HA epitope tags were cloned into
both cDNAs by PCR. Full-length BRAF cDNAs were subsequently cloned into
pBABE.puro.
Cell Culture and Transfection. WM793 melanoma cells were derived
from a vertical growth phase tumor as described previously (10), and HT1080
and HEK cells were obtained from American Type Culture Collection. Cells
were cultured under standard conditions (37°C in humidified atmosphere
containing 5%CO
2
) and grown in DMEM supplemented with 25 mM HEPES
(pH 7.4), 10% FCS, penicillin (100 units/ml), and streptomycin (100
g/ml).
To achieve transient suppression of gene expression, cells were plated in
six-well dishes at 5060% confluency and transfected with 5
g of duplex
RNA plus 6
l of OLIGOFECTAMINE (Life Technologies, Inc., Carlsbad,
CA) per the manufacturer’s recommendations and as described (8, 9).
The specificity of the targeting sequences was determined by transient
cotransfection of HEK cells with pBABE.puro.HA-tagged BRAF or
pBABE.puro.HA-tagged BRAF
V599E
and shRNA vectors (11). For stable trans
-
fection experiments, cells were plated at 5080% confluency in 100-mm
dishes and transfected with 4
g of plasmid DNA and 12
l of Fugene 6
(Roche, Indianapolis, IN) per the manufacturer’s instructions. Twenty-four h
after transfection, cells were selected in media containing 2
g/ml Puromycin
for 60–72 h and then collected for biochemical and cellular assays.
Immunoblotting. Adherent cells were washed with ice-cold PBS and lysed
and scraped in boiling SDS lysis buffer (10 m
M Tris, 1% SDS, 50 mM NaF, and
1m
M VO4). Lysates were boiled for 5 min, the DNA was sheared, and
insoluble debris was removed by microcentrifugation (14,000 rpm for 10 min).
Received 6/2/03; accepted 7/9/03.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact.
1
Supported in part by NIH Grant R25-CA87812 (S. R. H.), the McCabe Foundation
(D. A. T.), the Abramson Cancer Center of the University of Pennsylvania Pilot Projects
Program and Grant IRG-78-002-26 from the American Cancer Society (D. A. T.), The
Mary L. Smith Charitable Lead Trust (D. A. T.), and NIH Grants CA-25874, CA-47159,
CA-76674, and CA-10815 (M. H.).
2
To whom requests for reprints should be addressed, at University of Pennsylvania,
Department of Medicine, Philadelphia, PA 19104. E-mail: tuvesond@mail.med.
upenn.edu.
3
The abbreviations used are: MEK, mitogen-activated protein kinase kinase; ERK,
extracellular signal-regulated kinase; siRNA, small interfering RNA; HEK, human
embryonic kidney; HA, hemagglutinin; shRNA, short hairpin RNA; MAPK, mitogen-
activated protein kinase; RNAi, RNA interference; TBS, Tris-buffered saline.
5198
Protein concentrations were determined with bicinchoninic acid (Pierce, Rock-
ford, IL). Samples (15
g of total protein per lane) were resolved by reducing
SDS-PAGE and transferred to Immobilon-P polyvinylidene difluoride mem-
branes (Millipore, Bedford, MA). Membranes were blocked and incubated
with primary antibodies in TBS [150 mM Tris-HCL (pH 8.0) and 150 mM
NaCl] 3% BSA for antiphospho antibodies or 5% nonfat dry milk/TBS for
other antibodies. The membranes were subsequently washed (TBS/0.1%
Tween 20), incubated with horseradish peroxidase-conjugated secondary an-
tibodies, and washed again before being processed with enhanced chemilumi-
nescence plus (Amersham Biosciences, Little Chalfont, United Kingdom).
Membranes were probed sequentially for the indicated proteins after washing
in stripping buffer [50 mM Glycine (pH 2.5) and 0.05% Tween 20] for 15 min
at 55°C. Primary antibodies were procured from the following sources: an-
tiphosphorylated and total MEK (Cell Signaling, Beverly, MA), anti-HA
(Sigma, St. Louis, MO), and anti-Lamin A/C (Vector Laboratories, Burl-
ingame, CA) and antibodies against actin, B-Raf, and C-Raf were all obtained
from Santa Cruz Biotechnology (Santa Cruz, CA). Horseradish peroxidase-
conjugated secondary antibodies were from Jackson Immunoresearch (West
Grove, PA).
Proliferation, Apoptosis, and Transformation Assays. After selection,
shRNA-transfected cells were plated onto glass coverslips in media. The next
day, cells were incubated with 1 mM BrdUrd (Sigma) for 4 h, and positive
nuclei detected with anti-BrdUrd FITC per manufacturers instructions (Roche,
Indianapolis, IN). Apoptosis was detected with the In Situ Cell Death Detec-
tion kit per the manufacturers instructions (Roche). Three high-powered fields
were counted manually to determine the percentage of cells in S phase and the
degree of apoptosis, respectively. Nuclear staining was detected with 4,6-
diamidino-2-phenylindole (Sigma). Soft agar assays were performed by plating
50,000 cells/60-mm dish in 0.34% agar/media suspension over a solidified
0.5% agar layer. Dishes were replenished every 57 days.
Results
The recent development of facile methods for both transient and
stable suppression of gene expression by RNAi has provided powerful
tools for the study of mammalian cell genetics (8, 11, 12). These
methods exploit a conserved biological response to short duplex RNA
that results in post-transcriptional gene silencing (13). To evaluate the
role of BRAF expression in human melanoma cells, we generated a
series of stably expressing vectors against discrete regions of the
human BRAF coding sequence (Fig. 1A). We then tested the speci-
ficity of these reagents in HEK cells that had been transiently cotrans-
fected with HA epitope-tagged wild-type and mutant BRAF vectors
(Fig. 1B). Targeting sequences common to both wild-type and mutant
alleles (Com-1, 2, 3, and 4) caused various degrees of knockdown of
BRAF expression in HEK cells as demonstrated by immunoblots
against the expressed protein, with Com-4 being the most effective. A
mutant-specific vector (Mu-A) was also created that essentially com-
pletely abolished BRAF
V599E
expression while keeping wild-type
BRAF expression intact.
After establishing the efficacy and specificity of various RNAi
constructs described above, we designed corresponding duplex siRNA
species to study the effects of endogenous BRAF knockdown in
melanoma cells because of the higher transfection efficiencies achiev-
able by this method and the ability to obviate the need for antibiotic
selection. WM793 human melanoma cells were derived from a ver-
tical growth phase tumor (10) and have been shown to harbor the
BRAF
V599E
allele (14). Com-4 and Mu-A siRNA significantly inhib
-
ited BRAF expression and its attendant downstream signaling as
measured by phosphorylated MEK levels, although these effects were
relatively short lived and reversed by 7296 h after transfection (Fig.
2). Transfection of siRNA against Lamin A/C as a control revealed
efficient and more durable suppression of target expression, reflecting
either greater efficacy or stability of this transfected RNA duplex or
perhaps lower endogenous levels of lamin mRNA synthesis.
Despite the marked biochemical sequelae to BRAF knockdown by
siRNA, the effects were transient and rapidly reversible, and there were
no overt phenotypic consequences. Thus, to study the potential biological
consequences of more durable perturbations of these pathways, we used
our panel of shRNA vectors to stably knock down BRAF in melanoma
cells. Transfection of Com-4 and Mu-A shRNA vectors into WM793
cells effectively and stably suppressed BRAF expression and MEK phos-
phorylation (Fig. 3A). Cells transfected with control vector had no dis-
cernible effect on these parameters. On the other hand, although it was
possible to stably suppress BRAF expression by Com-4 in HT1080
human fibrosarcoma cells, no corresponding inhibition of MEK phos-
phorylation was observed, indicating that these cells activate the MAPK
pathway by another mechanism. As expected, transfection with Mu-A
had no discernible effect on B-Raf levels because these cells do not
contain the BRAF
V599E
mutation.
Several cellular consequences to stable BRAF suppression were
readily apparent. First, the morphology of WM793 cells changed
dramatically, becoming much larger and flatter and also less refractile
(Fig. 3B). In addition, far fewer WM793 cells were recovered after
Fig. 1. Design and characterization of vector-based RNAi of human BRAF. A, sche-
matic representation of human BRAF cDNA demonstrating sites selected for targeting. B,
efficacy of shRNA vectors in suppressing BRAF and BRAF
V599E
expression. Subconfluent
adherent HEK cells were either not transfected () or cotransfected with pBABE.pu-
ro.HA-BRAF (left panels) or pBABE.puro.HA-BRAF
V599E
(right panels), and the series of
shRNA plasmid constructs were directed against BRAF. After transfection (4872 h),
whole cell extracts were prepared and analyzed for HA expression; actin was used as a
loading control. This experiment was performed three times with similar results.
Fig. 2. Transient suppression of wild-type and mutant BRAF by siRNA in WM793
cells. Subconfluent cell cultures were transfected with siRNA directed against Lamin A/C
(L), BRAF (4), and BRAF
V599E
(A). Whole cell lysates were then prepared at 24-, 48-, 72-,
and 96-h post-transfection and analyzed for specific protein expression by immunoblot-
ting with the indicated antibodies. The position of the M
r
45,000 phospho-MEK 1/2
proteins is indicated; the identity of the faster migrating band is unknown. This experiment
was performed three times with similar results.
5199
BRAF
V599E
SUPPRESSION IN HUMAN MELANOMA
transfection with Com-4, as compared with control vector or Mu-A,
suggesting that wild-type B-Raf function may also be important for
the viability of these cells. No appreciable differences in morphology
or cell number were noted in parallel transfections of HT1080 cells
(Fig. 3B). In addition, Com-4 and Mu-A-transfected WM793 cells
exhibited markedly lower proliferative rates; the percentage of cells in
S phase was 4 / 2% and 0.8 / 1%, respectively, as compared
with 19 / 2% for cells transfected with empty vector (Fig. 3C).
The proliferation of HT1080 cells was not affected by transfection
with any of these vectors, again despite achieving significant knock-
down of B-Raf levels (Fig. 3C). WM793 cells also exhibited increased
levels of apoptosis after stable suppression of BRAF, whereas human
fibrosarcoma cells again remained unaffected by similar manipula-
tions (Fig. 3D).
These results demonstrated that BRAF-dependent signaling was
necessary for the optimal proliferation and survival of human mela-
noma WM793 cells and dispensable for human fibrosarcoma cells.
We wondered whether these effects were specific for the BRAF family
member of the Raf kinases or extended to the heretofore more exten-
sively studied homologue, CRAF. We therefore generated CRAF-
specific duplex siRNA species and stably expressing shRNA vectors
and tested their abilities to suppress CRAF expression and inhibit
downstream phosphorylation of MEK. As shown in Fig. 4A, knock-
down of C-Raf protein levels by siRNA followed a slower time course
than that of B-Raf and remained more durably suppressed. Notably,
however, there appeared to be no effect on MEK phosphorylation
despite nearly complete suppression of CRAF. Thus, at least in
WM793 melanoma cells, CRAF appears not to be required for MEK
activation. A stably expressing shRNA vector directed against the
same sequence similarly knocked down CRAF expression with no
attendant affect on MEK phosphorylation (Fig. 4B).
Finally, the ability to manifest anchorage-independent growth, an
established feature of cellular transformation, was assessed in mela-
noma cells after the knockdown of BRAF, BRAF
V599E
, or CRAF.
WM793 cells transfected with empty vector readily formed colonies
in soft agar (Fig. 4C). Cells in which C-Raf levels had been stably
knocked down formed colonies almost as readily. Transfection with
either Com-4 or Mu-A, however, essentially completely abrogated the
ability of these cells to manifest anchorage-independent growth.
Therefore, BRAF is uniquely required for cellular transformation in
WM793 cells.
Discussion
Oncogenesis is generally viewed as a multistep process character-
ized by the progressive acquisition of genetic mutations and func-
tional capabilities (15). The hope for curative therapies lies in the
proposition that key genetic events exist which represent unique
points of vulnerability for cancer cells. Indeed, despite the complexity
of genetic and epigenetic alterations in cancer cells and their micro-
environment, recent evidence demonstrates that the specific inhibition
of one, or perhaps a few, critical pathways in tumor cells may be
sufficient to kill them and provide significant clinical benefit. As
examples, treatment of patients with stable phase chronic myeloge-
Fig. 3. Effects of stable suppression of endogenous BRAF and BRAF
V599E
in WM793 and HT1080 cells. Subconfluent cultures of WM793 or HT1080 cells were transfected with
pSUPER.retro (vector), shRNA directed against BRAF (Com-4), or shRNA directed against BRAF
V599E
(Mu-A). After selection in puromycin, the biochemical and biological properties
of these cells were assessed. A, immunoblot analysis of B-Raf, activated MEK (p-MEK), total MEK (t-MEK), and actin levels. The positions of the M
r
45,000 phospho- and total-MEK
1/2 proteins are indicated. B, phase contrast microscopy of cells in culture (magnification: 100). C, proliferative index as measured by BrdUrd incorporation. D, degree of apoptosis
as assessed by TUNEL reactivity. BrdUrd and TUNEL data are expressed as the means / SD of direct counting of three high-powered fields. These experiments were performed
twice with similar results.
5200
BRAF
V599E
SUPPRESSION IN HUMAN MELANOMA
nous leukemia or advanced gastrointestinal stromal tumors with ima-
tinib mesylate, a small molecule inhibitor of the ABL and KIT tyrosine
kinases, respectively, induces dramatic remissions with minimal tox-
icity (16, 17). Importantly, the responses of chronic myelogenous
leukemia and gastrointestinal stromal tumor patients to imatinib cor-
relates with the drugs ability to inhibit the kinase activities of Bcr-
Abl and mutant c-kit (18, 19), respectively. These results suggest that
essential pathways may exist in other malignancies and that biochem-
ical confirmation of the effectiveness of molecularly targeted thera-
pies may be predictive of clinical efficacy.
Our findings suggest that the BRAF
V599E
mutation commonly
found in malignant melanomas may represent a therapeutic target
analogous to BCR-ABL and KIT. We have demonstrated here that
knockdown of BRAF expression and inhibition of downstream sig-
naling in WM793 human melanoma cells causes growth arrest and
promotes apoptosis under adherent conditions, and prevents colony
formation in suspension. These observations have been preliminarily
extended to a second melanoma cell line known to contain the
BRAF
V599E
mutation (data not shown). These effects were specific to
BRAF, as suppression of CRAF failed to inhibit downstream phos-
phorylation of MEK and did not appreciably alter the biological
properties of these cells. Moreover, these effects were specific to
melanoma cells, because human fibrosarcoma cells were impervious
to suppression of BRAF expression.
Fig. 4. Effects of transient and stable suppression of BRAF or CRAF expression in WM793 cells. In A, WM793 cells were transfected with siRNA directed against Lamin A/C (L),
BRAF (4), BRAF
V599E
(A), and CRAF (C) and analyzed for specific protein expression at 24, 48, and 96 h post-transfection. In B, WM793 cells were stably transfected with
pSUPER.retro (vector) or shRNA directed against CRAF (C-1), subjected to antibiotic selection, and assessed for knockdown of C-Raf, activated MEK (p-MEK), total MEK (t-MEK),
and actin levels. The positions of the M
r
45,000 phospho- and total-MEK 1/2 proteins are indicated. In C, WM793 cells were stably transfected with pSUPER.retro, Com-4 shRNA
against BRAF, C-1 shRNA against C-Raf, or Mu-A shRNA against BRAF
V599E
and plated onto semisolid media. Colonies were counted and photographed after 30 days of growth
(magnification: 40). These results are representative of two independent experiments.
5201
BRAF
V599E
SUPPRESSION IN HUMAN MELANOMA
Currently, a Raf kinase inhibitor, BAY 439006 (20), is undergoing
worldwide clinical evaluation in Phase I and II trials in patients with
a variety of malignancies, including melanoma. However, BAY 43
9006 inhibits both B-Raf and C-Raf kinase activities,
4
and any ben-
eficial or adverse effects of treatment may therefore result from
simultaneous inhibition of both kinases. Our results suggest that
targeted inhibition of B-Raf specifically in such tumors may be
equally efficacious and perhaps associated with less toxicity.
That CRAF expression was dispensable for the transformed pheno-
type in human melanoma cells was somewhat surprising. As the first
of the three Raf family isoforms identified, a large body of evidence
exists exploring the transforming properties of CRAF in mammalian
cell systems. These properties, however, are exquisitely dependent on
cellular context (7); for example, although a constitutively active form
of CRAF can readily transform NIH 3T3 cells, it is unable to do so in
RIE-1 cells (21). More recent experiments involving targeted disrup-
tion and mutation of Raf isoforms in mice implicate B-Raf as the more
potent activator of MEK in many cell and tissue types (22). The
minimal effects on transformation of CRAF suppression in the mel-
anoma cells studied here suggest that this isoform may not always be
the predominant effector of MAPK signaling in human cells either.
In summary, we find that suppression of BRAF
V599E
in WM793
human melanoma cells abrogates their transformed phenotype and
conclude, therefore, that agents that specifically inhibit activated
BRAF, and not CRAF, might be particularly efficacious in melanomas,
and perhaps other tumor types, that harbor activating mutations in this
proto-oncogene.
References
1. Jemal, A., Murray, T., Samuels, A., Ghafoor, A., Ward, E., and Thun, M. J. Cancer
statistics, 2003. CA Cancer J. Clin., 53: 526, 2003.
2. Jemal, A., Devesa, S. S., Hartge, P., and Tucker, M. A. Recent trends in cutaneous
melanoma incidence among whites in the United States. J. Natl. Cancer Inst. (Be-
thesda), 93: 678683, 2001.
3. Balch, C. M., Buzaid, A. C., Soong, S. J., Atkins, M. B., Cascinelli, N., Coit, D. G.,
Fleming, I. D., Gershenwald, J. E., Houghton, A., Jr., Kirkwood, J. M., McMasters,
K. M., Mihm, M. F., Morton, D. L., Reintgen, D. S., Ross, M. I., Sober, A.,
Thompson, J. A., and Thompson, J. F. Final version of the American Joint Committee
on Cancer staging system for cutaneous melanoma. J. Clin. Oncol., 19: 36353648,
2001.
4. Brose, M. S., Volpe, P., Feldman, M., Kumar, M., Rishi, I., Gerrero, R., Einhorn, E.,
Herlyn, M., Minna, J., Nicholson, A., Roth, J. A., Albelda, S. M., Davies, H., Cox, C.,
Brignell, G., Stephens, P., Futreal, P. A., Wooster, R., Stratton, M. R., and Weber,
B. L. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res.,
62: 69977000, 2002.
5. Davies, H., Bignell, G. R., Cox, C., Stephens, P., Edkins, S., Clegg, S., Teague, J.,
Woffendin, H., Garnett, M. J., Bottomley, W., Davis, N., Dicks, E., Ewing, R., Floyd,
Y., Gray, K., Hall, S., Hawes, R., Hughes, J., Kosmidou, V., Menzies, A., Mould, C.,
Parker, A., Stevens, C., Watt, S., Hooper, S., Wilson, R., Jayatilake, H., Gusterson,
B. A., Cooper, C., Shipley, J., Hargrave, D., Pritchard-Jones, K., Maitland, N.,
Chenevix-Trench, G., Riggins, G. J., Bigner, D. D., Palmieri, G., Cossu, A., Flanagan,
A., Nicholson, A., Ho, J. W., Leung, S. Y., Yuen, S. T., Weber, B. L., Seigler, H. F.,
Darrow, T. L., Paterson, H., Marais, R., Marshall, C. J., Wooster, R., Stratton, M. R.,
and Futreal, P. A. Mutations of the BRAF gene in human cancer. Nature (Lond.), 417:
949954, 2002.
6. Pollock, P. M., Harper, U. L., Hansen, K. S., Yudt, L. M., Stark, M., Robbins, C. M.,
Moses, T. Y., Hostetter, G., Wagner, U., Kakareka, J., Salem, G., Pohida, T., Heenan,
P., Duray, P., Kallioniemi, O., Hayward, N. K., Trent, J. M., and Meltzer, P. S. High
frequency of BRAF mutations in nevi. Nat. Genet., 33: 1920, 2003.
7. Hagemann, C., and Rapp, U. R. Isotype-specific functions of Raf kinases. Exp. Cell
Res., 253: 3446, 1999.
8. Elbashir, S. M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., and Tuschl, T.
Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian
cells. Nature (Lond.), 411: 494498, 2001.
9. Elbashir, S. M., Harborth, J., Weber, K., and Tuschl, T. Analysis of gene function in
somatic mammalian cells using small interfering RNAs. Methods, 26: 199213,
2002.
10. Satyamoorthy, K., DeJesus, E., Linnenbach, A. J., Kraj, B., Kornreich, D. L., Rendle,
S., Elder, D. E., and Herlyn, M. Melanoma cell lines from different stages of
progression and their biological and molecular analyses. Melanoma Res., 7 (Suppl.
2): S35S42, 1997.
11. Brummelkamp, T. R., Bernards, R., and Agami, R. A system for stable expression of
short interfering RNAs in mammalian cells. Science, 296: 550553, 2002.
12. Paddison, P. J., Caudy, A. A., and Hannon, G. J. Stable suppression of gene
expression by RNAi in mammalian cells. Proc. Natl. Acad. Sci. USA, 99: 14431448,
2002.
13. Hannon, G. J. RNA interference. Nature (Lond.), 418: 244251, 2002.
14. Satyamoorthy, K., Li, G., Gerrero, M. R., Brose, M. S., Volpe, P., Weber, B. L., Van
Belle, P., Elder, D. E., and Herlyn, M. Constitutive mitogen-activated protein kinase
activation in melanoma is mediated by both BRAF mutations and autocrine growth
factor stimulation. Cancer Res., 63: 756759, 2003.
15. Hanahan, D., and Weinberg, R. A. The hallmarks of cancer. Cell, 100: 5770, 2000.
16. Druker, B. J., Talpaz, M., Resta, D. J., Peng, B., Buchdunger, E., Ford, J. M., Lydon,
N. B., Kantarjian, H., Capdeville, R., Ohno-Jones, S., and Sawyers, C. L. Efficacy and
safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid
leukemia. N. Engl. J. Med., 344: 10311037, 2001.
17. Demetri, G. D., von Mehren, M., Blanke, C. D., Van den Abbeele, A. D., Eisenberg,
B., Roberts, P. J., Heinrich, M. C., Tuveson, D. A., Singer, S., Janicek, M., Fletcher,
J. A., Silverman, S. G., Silberman, S. L., Capdeville, R., Kiese, B., Peng, B.,
Dimitrijevic, S., Druker, B. J., Corless, C., Fletcher, C. D., and Joensuu, H. Efficacy
and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N. Eng.
J. Med., 347: 472480, 2002.
18. Gorre, M. E., Mohammed, M., Ellwood, K., Hsu, N., Paquette, R., Rao, P. N., and
Sawyers, C. L. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL
gene mutation or amplification. Science (Wash. DC), 293: 876880, 2001.
19. Tuveson, D. A., Willis, N. A., Jacks, T., Griffin, J. D., Singer, S., Fletcher, C. D.,
Fletcher, J. A., and Demetri, G. D. STI571 inactivation of the gastrointestinal stromal
tumor c-KIT oncoprotein: biological and clinical implications. Oncogene, 20: 5054
5058, 2001.
20. Lyons, J. F., Wilhelm, S., Hibner, B., and Bollag, G. Discovery of a novel Raf kinase
inhibitor. Endocrine-Related Cancer, 8: 219225, 2001.
21. Oldham, S. M., Clark, G. J., Gangarosa, L. M., Coffey, R. J., Jr., and Der, C. J.
Activation of the Raf-1/MAP kinase cascade is not sufficient for Ras transformation
of RIE-1 epithelial cells. Proc. Natl. Acad. Sci. USA, 93: 69246928, 1996.
22. Mercer, K. E., and Pritchard, C. A. Raf proteins and cancer: B-Raf is identified as a
mutational target. Biochim. Biophys. Acta, 1653: 2540, 2003.
4
G. Bollag, personal communication.
5202
BRAF
V599E
SUPPRESSION IN HUMAN MELANOMA
... BRAF mutants are common in melanomas, thyroid and colorectal carcinomas, and other types of tumors. Proliferation of these tumors is sensitive to ERK inhibition by MEK or RAF inhibitors and genetic ablation of BRAF V600E allele [3][4][5]. This is consistent with the substantial clinical benefit obtained with BRAF inhibitors in BRAF V600E mutant melanomas [3,[6][7][8]. ...
Preprint
Full-text available
BRAF V600E mutation occurs in 46% of melanomas and drives high levels of ERK activity and ERK-dependent proliferation. However, BRAF V600E is insufficient to drive melanoma in GEMM models, and 82% of human benign nevi harbor BRAF V600E mutations. We show here that BRAF V600E inhibits mesenchymal migration by causing feedback inhibition of RAC1 activity. ERK pathway inhibition induces RAC1 activation and restores migration and invasion. In cells with BRAF V600E , mutant RAC1, overexpression of PREX1, PREX2, or PTEN inactivation restore RAC1 activity and cell motility. Together, these lesions occur in 48% of BRAF V600E melanomas. Thus, although BRAF V600E activation of ERK deregulates cell proliferation, it prevents full malignant transformation by causing feedback inhibition of cell migration. Secondary mutations are, therefore, required for tumorigenesis. One mechanism underlying tumor evolution may be the selection of lesions that rescue the deleterious effects of oncogenic drivers. Statement of significance BRAF V600E activation of ERK causes feedback inhibition of cell migration and invasion and thus blocks tumorigenesis. Secondary genetic lesions are required to rescue these processes and enable tumor development. Thus, oncogenic feedback can shape the details of tumor progression and, in doing so, selects for new mutations that may be therapeutic targets.
... The expression of bacterial recombinase Cre results well into the withdrawal of the LSL cassette, expression of the mutant K-rasG12D from its endogenous locus, and thus the initiation of the Ras pathway [40]. Usually, all pancreatic cells are delivered from pancreatic progenitors [41]. By crossing PDX-1-Cre or p48-Cre mice to K-rasLSL G12D mice, the outcome from this crossing is a mouse model known as KC [42]. ...
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) is among the dangerous human cancers, is the 10th highly prevalent cancer, and the fourth sole cause of cancer-related mortality in the United States of America. Notwithstanding the significant commitment, the forecast for people with this burden continues to have a five-year survival rate of just 4-6%. The most critical altered genes within PDAC consist of K-ras the proto-oncogene which is usually mutationally activated above 90% cases and tumor suppressors likeTrp53 are altered at 55%. To face the burden of pancreatic ductal adenocarcinoma, a variety of genetically engineered pancreatic cancer mice models have been created over the last past years. These models have distinctive features and are not all appropriate for preclinical studies. In this review, we focus on differences between two mice models K-rasLSL.G12D/+;Pdx-1-Cre(KC) and K-rasLSL.G12D/+; Trp53R172H/+; Pdx-1-Cre(KPC) in terms of their modeling biology and their clinical relevance.
... Increasing evidence suggests that molecular profiling will add additional information supporting to the staging and prognosis prediction of melanoma patients. Hence, attributing to an improved cognition of disease at molecular level, improvements in targeting therapies for metastatic melanoma patients are obtained [29][30][31][32][33]. Nevertheless, biomarkers for the diagnosis, prognosis prediction, and guidance of treatment for melanoma patients are still lacking. ...
Article
Full-text available
Melanoma is a malignant tumor that originates in melanocytes of the skin or mucous membrane, which has a high mortality rate and worse prognosis. Therefore, perspective prognosis evaluation seems more important for patients’ treatment. Gene expression profiles of melanoma were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, respectively. 130 consistent differentially expressed genes (DEGs) were identified between melanoma and nevus tissues from two GEO cohorts. Prognostic genes were identified by univariate analysis, and 20 of them were regarded to be associated with the recurrence-free survival (RFS) of melanoma patients. Then, the LASSO Cox regression analysis chose seven of them to establish a seven-DEG-based RFS predicting signature. We demonstrated that this model was more powerful to predict RFS risk than other individual clinical features and was able to independently predict the RFS outcomes in different subsets of patients. We attempted to search for the underlying mechanisms by analyzing the coexpression genes of the seven candidates, and the pathway enrichment analyses indicated that immune response-related pathways might play a critical role in melanoma progression. Finally, we establish a robust seven-DEG-based RFS predicting signature, which will facilitate the personalized treatment of melanoma patients.
... Среди терапевтических мишеней сигнальный механизм митоген-активированных протеинкиназ (МАРК) заслуживает более пристального внимания, так как этот путь активируется в большинстве случаев меланомы кожи [5]. Мутагенная активация МАР-киназного пути наблюдается более чем в 90% случаев меланомы, и наиболее часто активация происходит через мутации генов NRAS или BRAF [5,6]. Мутация гена BRAF ведет к стабильной активации сигнального механизма МАРК и также выявляется в доброкачественных меланоцитарных образованиях (невусах), что доказывает роль BRAF в качестве активатора клеточной пролиферации, но не злокачественной трансформации [7]. ...
Article
Full-text available
Skin melanoma is an aggressive malignant neoplasm. Mutation of the BRAF gene, BRAF V600E, is observed in more than 70% of melanoma cases. Assessment of the BRAF status in patients suffering from skin melanoma is an important stage in patient screening for selection of further therapy. To reveal the mutation of BRAF V600E, 27 histology samples taken from patients with skin melanoma residing in the Krasnodar territory were examined by means of defining the Restriction Fragment Length Polymorphism (PFLP assay). Our studies revealed the BRAF V600E mutation in 26 of 27 histology samples of skin melanoma. It was shown that the mutation is not associated with any sex-related, age-related, clinical and pathological features of the tumor.
Article
Full-text available
The RAS/mitogen-activated protein kinase (MAPK) signaling cascade is commonly dysregulated in human malignancies by processes driven by RAS or RAF oncogenes. Among the members of the RAF kinase family, CRAF plays an important role in the RAS-MAPK signaling pathway, as well as in the progression of cancer. Recent research has provided evidence implicating the role of CRAF in the physiological regulation and the resistance to BRAF inhibitors through MAPK-dependent and MAPK-independent mechanisms. Nevertheless, the effectiveness of solely targeting CRAF kinase activity remains controversial. Moreover, the kinase-independent function of CRAF may be essential for lung cancers with KRAS mutations. It is imperative to develop strategies to enhance efficacy and minimize toxicity in tumors driven by RAS or RAF oncogenes. The review investigates CRAF alterations observed in cancers and unravels the distinct roles of CRAF in cancers propelled by diverse oncogenes. This review also seeks to summarize CRAF-interacting proteins and delineate CRAF's regulation across various cancer hallmarks. Additionally, we discuss recent advances in pan-RAF inhibitors and their combination with other therapeutic approaches to improve treatment outcomes and minimize adverse effects in patients with RAF/RAS-mutant tumors. By providing a comprehensive understanding of the multifaceted role of CRAF in cancers and highlighting the latest developments in RAF inhibitor therapies, we endeavor to identify synergistic targets and elucidate resistance pathways, setting the stage for more robust and safer combination strategies for cancer treatment.
Article
Full-text available
Simple Summary Mutational hotspots have gained importance as oncological biomarkers in recent years because of their potential as predictors of clinical outcomes and/or therapeutic targets. In addition, they are easily detectable in clinical samples via Sanger or next-generation sequencing (NGS). The role of these genetic defects is less clear in pituitary neuroendocrine tumors (PitNETs), even though the most common genetic drivers of these neoplasms are located within mutational hotspots. Indeed, hotspots in six different genes are of particular importance in this context. Two of them, USP48 and SF3B1, represent very recent and infrequent genetic associations; thus, their clinical relevance remains unclear. For two other genes, GNAS and USP8, discrepancies exist among studies regarding their associated phenotypes. Finally, the phenotypes associated with BRAF and DICER1 are well defined in other settings, but not yet in sporadic PitNETs. Additional studies are required to assess the potential of these molecular alterations as druggable targets in PitNETs. Abstract The most common genetic drivers of pituitary neuroendocrine tumors (PitNETs) lie within mutational hotspots, which are genomic regions where variants tend to cluster. Some of these hotspot defects are unique to PitNETs, while others are associated with additional neoplasms. Hotspot variants in GNAS and USP8 are the most common genetic causes of acromegaly and Cushing’s disease, respectively. Although it has been proposed that these genetic defects could define specific clinical phenotypes, results are highly variable among studies. In contrast, DICER1 hotspot variants are associated with a familial syndrome of cancer predisposition, and only exceptionally occur as somatic changes. A small number of non-USP8-driven corticotropinomas are due to somatic hotspot variants in USP48 or BRAF; the latter is a well-known mutational hotspot in cancer. Finally, somatic variants affecting a hotspot in SF3B1 have been associated with multiple cancers and, more recently, with prolactinomas. Since the associations of BRAF, USP48, and SF3B1 hotspot variants with PitNETs are very recent, their effects on clinical phenotypes are still unknown. Further research is required to fully define the role of these genetic defects as disease biomarkers and therapeutic targets.
Chapter
Several targeted therapies are currently used in everyday clinical practice for the treatment of Stage IIIC/IV melanoma. These medications target the mitogen-activated protein kinase (MAPK) pathway. B-Raf proto-oncogene (BRAF) inhibitors such as vemurafenib, dabrafenib, and encorafenib are indicated as first-line therapies in BRAFV600E mutation positive melanomas and are administered in combination with the mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors trametinib, cobimetinib, and binimetinib, respectively. Combination regimens have been associated with fast responses, acceptable efficacy, and improved duration of response compared to BRAF inhibitor monotherapy, somewhat limiting the drug resistance problem that is frequently encountered with these drugs. Adverse events associated with their use include a variety of cutaneous, gastrointestinal, cardiovascular, and ocular symptoms, among others.
Article
Melanoma had long been considered to be particularly addressable with immunotherapy, but that reputation was built on modestly effective cytokine‐based immunotherapy. CTLA‐4 antibody therapy reinforced this legacy, but PD‐1 antibodies transformed the melanoma treatment landscape and lead the way for immunotherapy to become standard treatment for more than half of the advanced cancer population. BRAF mutations were discovered in 8% of all cancer and nearly 50% of melanomas. Successful development of BRAF inhibitors and BRAF/MEK combination therapy in melanoma preceded regulatory approval across all cancer types. No cancer type saw outcomes improved by the same margin as melanoma in the decade of the 2010s.
Article
Targeted drug delivery (TDD) is the selective delivery of a therapeutic agent specifically to the site of action to avoid adverse effects and systemic toxicity and to reduce the dose required. Ligand TDD or active TDD involves using a ligand-drug conjugate comprising a targeting ligand linked to an active drug moiety that can either be free or encapsulated within a nanocarrier (NC). Aptamers are single-stranded oligonucleotides that bind to specific biomacromolecules because of their 3D conformation. Nanobodies are the variable domains of unique heavy chain-only antibodies (HcAbs) produced by animals of the Camelidae family. Both these types of ligand are smaller than antibodies and have been used to efficiently target drugs to particular tissues or cells. In this review, we describe the applications of aptamers and nanobodies as ligands for TDD, their advantages and disadvantages compared with antibodies, and the various modalities for targeting cancers using these ligands. Teaser: Aptamers and nanobodies are macromolecular ligands that can actively chaperone drug molecules to particular cancerous cells or tissues in the body to target their pharmacological effects and improve their therapeutic index and safety.
Article
Full-text available
We discuss the biology of Ras signal transduction and the epidemiology of ras mutations in association with disease as a background for the development of a Raf kinase inhibitor, BAY 43-9006. Knowledge of Ras effector pathways has permitted genetic validation of numerous targets involved in the Ras signaling cascade. A key Ras effector pathway involves the kinase cascade RAF/MEK/ ERK (MEK: MAP/ERK kinase; ERK: extracellular signal related kinase). Indeed, we present studies of cell lines stably expressing mutant MEK constructs, which point to Raf kinase as a target for therapeutics with selective anti-tumor activity. Finally, a small molecule drug discovery program based on inhibition of Raf kinase activity is outlined and the initial pre-clinical development process of the Raf kinase inhibitor BAY 43-9006 is discussed.
Article
Full-text available
The potent transforming activity of membrane-targeted Raf-1 (Raf-CAAX) suggests that Ras transformation is triggered primarily by a Ras-mediated translocation of Raf-1 to the plasma membrane. However, whereas constitutively activated mutants of Ras [H-Ras(61L) and K-Ras4B(12V)] and Raf-1 (DeltaRaf-22W and Raf-CAAX) caused indistinguishable morphologic and growth (in soft agar and nude mice) transformation of NIH 3T3 fibroblasts, only mutant Ras caused morphologic transformation of RIE-1 rat intestinal cells. Furthermore, only mutant Ras-expressing RIE-1 cells formed colonies in soft agar and developed rapid and progressive tumors in nude mice. We also observed that activated Ras, but not Raf-1, caused transformation of IEC-6 rat intestinal and MCF-10A human mammary epithelial cells. Although both Ras- and DeltaRaf-22W-expressing RIE-1 cells showed elevated Raf-1 and mitogen-activated protein (MAP) kinase activities, only Ras-transformed cells produced secreted factors that promoted RIE-1 transformation. Incubation of untransformed RIE-1 cells in the presence of conditioned medium from Ras-expressing, but not DeltaRaf-22W-expressing, cells caused a rapid and stable morphologic transformation that was indistinguishable from the morphology of Ras-transformed RIE-1 cells. Thus, induction of an autocrine growth mechanism may distinguish the transforming actions of Ras and Raf. In summary, our observations demonstrate that oncogenic Ras activation of the Raf/MAP kinase pathway alone is not sufficient for full tumorigenic transformation of RIE-1 epithelial cells. Thus, Raf-independent signaling events are essential for oncogenic Ras transformation of epithelial cells, but not fibroblasts.
Article
Full-text available
It is not yet clear whether increasing melanoma incidence is real or whether recent incidence trends mainly reflect improved diagnosis. To address this question, we examined the most recent melanoma incidence patterns among the white population stratified by sex, age, tumor stage, and tumor thickness by use of data from the Surveillance, Epidemiology, and End Results Program. We examined log-transformed age-specific rates for melanoma by 5-year age groups and time periods by year of diagnosis and birth cohort. Melanoma trends were further examined among broader age groups (<40 years, 40-59 years, and > or =60 years) by tumor stage and tumor thickness. Rates were age-adjusted to the 1970 U.S. standard population, and trends were tested by use of a two-sided Student's t test. Melanoma incidence increased in females born since the 1960s. From 1974-1975 through 1988-1989, upward trends for the incidence of localized tumors and downward trends for the incidence of distant-stage tumors occurred in the age group under 40 years. In the more recent time period, 1990-1991 through 1996-1997, age specific rates among females compared with males generally remained stable or declined more for distant-stage tumors and increased less for local-stage tumors. Thin tumors (<1 mm) increased statistically significantly in all age groups (P<.05 for all), except in men under age 40 years. In contrast, rates for thick tumors (> or =4 mm) increased statistically significantly (P =.0003) only in males aged 60 years and older. Melanoma incidence may well continue to rise in the United States, at least until the majority of the current population in the middle-age groups becomes the oldest population. The recent trends may reflect increased sunlight exposure.
Article
The biological and molecular characteristics of cell lines from metastatic melanomas have been extensively studied but less is known about cells from the biologically earliest stage of primary melanoma. The overall success rate of establishing permanent cell lines from such lesions is only 10% of that for biologically late primary or metastatic melanomas, although our laboratory now has eight cell lines available. The cells are immortal but show reduced or no proliferation in soft agar and immunodeficient mice when compared with primary melanomas from the biologically advanced vertical growth phase. Metastatic melanoma cell lines from patients with familial melanoma or xeroderma pigmentosum are biologically similar to those from patients with spontaneous melanomas. Irrespective of the malignant stages, deletions and mutations can occur in exons 1-3 of the p16INK4A gene. DNA fingerprinting was then employed to demonstrate the uniqueness of individual cell lines and to confirm the identity of cell lines derived from same patients. These cell lines are an excellent resource to investigate melanoma progression.
Article
The family of Raf-protein kinases consisting of A-Raf, B-Raf, and c-Raf-1 is involved in cellular processes which regulate proliferation, differentiation, and apoptosis. Cell-culture experiments and the knockout of individual Raf genes suggested that the three Raf isoforms have overlapping and unique regulatory functions. However, it is not known how these isotype-specific functions of Raf kinases occur in the cell. Published data suggest that Raf proteins might differ in the regulation of their activation as well as in their ability to connect to downstream signaling pathways. Since Raf is part of a multiprotein complex and protein-protein interactions are important for Raf signaling, we propose that isotype-specific functions can be achieved by isotype-restricted protein binding. Recently we were able to identify candidates for such Raf-isoform-specific interaction partners.
Article
We wish to thank Terry Schoop of Biomed Arts Associates, San Francisco, for preparation of the figures, Cori Bargmann and Zena Werb for insightful comments on the manuscript, and Normita Santore for editorial assistance. In addition, we are indebted to Joe Harford and Richard Klausner, who allowed us to adapt and expand their depiction of the cell signaling network, and we appreciate suggestions on signaling pathways from Randy Watnick, Brian Elenbas, Bill Lundberg, Dave Morgan, and Henry Bourne. R. A. W. is a Ludwig Foundation and American Cancer Society Professor of Biology. His work has been supported by the Department of the Army and the National Institutes of Health. D. H. acknowledges the support and encouragement of the National Cancer Institute. Editorial policy has rendered the citations illustrative but not comprehensive.
Article
BCR-ABL is a constitutively activated tyrosine kinase that causes chronic myeloid leukemia (CML). Since tyrosine kinase activity is essential to the transforming function of BCR-ABL, an inhibitor of the kinase could be an effective treatment for CML. We conducted a phase 1, dose-escalating trial of STI571 (formerly known as CGP 57148B), a specific inhibitor of the BCR-ABL tyrosine kinase. STI571 was administered orally to 83 patients with CML in the chronic phase in whom treatment with interferon alfa had failed. Patients were successively assigned to 1 of 14 doses ranging from 25 to 1000 mg per day. Adverse effects of STI571 were minimal; the most common were nausea, myalgias, edema, and diarrhea. A maximal tolerated dose was not identified. Complete hematologic responses were observed in 53 of 54 patients treated with daily doses of 300 mg or more and typically occurred in the first four weeks of therapy. Of the 54 patients treated with doses of 300 mg or more, cytogenetic responses occurred in 29, including 17 (31 percent of the 54 patients who received this dose) with major responses (0 to 35 percent of cells in metaphase positive for the Philadelphia chromosome); 7 of these patients had complete cytogenetic remissions. STI571 is well tolerated and has significant antileukemic activity in patients with CML in whom treatment with interferon alfa had failed. Our results provide evidence of the essential role of BCR-ABL tyrosine kinase activity in CML and demonstrate the potential for the development of anticancer drugs based on the specific molecular abnormality present in a human cancer.
Article
RNA interference (RNAi) is the process of sequence-specific, post-transcriptional gene silencing in animals and plants, initiated by double-stranded RNA (dsRNA) that is homologous in sequence to the silenced gene. The mediators of sequence-specific messenger RNA degradation are 21- and 22-nucleotide small interfering RNAs (siRNAs) generated by ribonuclease III cleavage from longer dsRNAs. Here we show that 21-nucleotide siRNA duplexes specifically suppress expression of endogenous and heterologous genes in different mammalian cell lines, including human embryonic kidney (293) and HeLa cells. Therefore, 21-nucleotide siRNA duplexes provide a new tool for studying gene function in mammalian cells and may eventually be used as gene-specific therapeutics.
Article
Clinical studies with the Abl tyrosine kinase inhibitor STI-571 in chronic myeloid leukemia demonstrate that many patients with advanced stage disease respond initially but then relapse. Through biochemical and molecular analysis of clinical material, we find that drug resistance is associated with the reactivation of BCR-ABL signal transduction in all cases examined. In six of nine patients, resistance was associated with a single amino acid substitution in a threonine residue of the Abl kinase domain known to form a critical hydrogen bond with the drug. This substitution of threonine with isoleucine was sufficient to confer STI-571 resistance in a reconstitution experiment. In three patients, resistance was associated with progressiveBCR-ABL gene amplification. These studies provide evidence that genetically complex cancers retain dependence on an initial oncogenic event and suggest a strategy for identifying inhibitors of STI-571 resistance.
Article
To revise the staging system for cutaneous melanoma under the auspices of the American Joint Committee on Cancer (AJCC). The prognostic factors analysis described in the companion publication (this issue), as well as evidence from the published literature, was used to assemble the tumor-node-metastasis criteria and stage grouping for the melanoma staging system. Major changes include (1) melanoma thickness and ulceration but not level of invasion to be used in the T category (except for T1 melanomas); (2) the number of metastatic lymph nodes rather than their gross dimensions and the delineation of clinically occult (ie, microscopic) versus clinically apparent (ie, macroscopic) nodal metastases to be used in the N category; (3) the site of distant metastases and the presence of elevated serum lactic dehydrogenase to be used in the M category; (4) an upstaging of all patients with stage I, II, and III disease when a primary melanoma is ulcerated; (5) a merging of satellite metastases around a primary melanoma and in-transit metastases into a single staging entity that is grouped into stage III disease; and (6) a new convention for defining clinical and pathologic staging so as to take into account the staging information gained from intraoperative lymphatic mapping and sentinel node biopsy. This revision will become official with publication of the sixth edition of the AJCC Cancer Staging Manual in the year 2002.