Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2008 Dec 2;58(8):1185–1194. doi: 10.1007/s00262-008-0623-1

Breast cancer cells expressing stem cell markers CD44+ CD24lo are eliminated by Numb-1 peptide-activated T cells

Takashi Mine 1,2,9,, Satoko Matsueda 1, Yufeng Li 1,6, Hiroshi Tokumitsu 3, Hui Gao 4, Cristopher Danes 5, Kwong-Kwok Wong 1, Xinhui Wang 7, Soldano Ferrone 7, Constantin G Ioannides 1,8
PMCID: PMC2726795  NIHMSID: NIHMS129775  PMID: 19048252

Abstract

Cancer stem cells (CSC) are resistant to chemo- and radiotherapy. To eliminate cells with phenotypic markers of CSC-like we characterized: (1) expression of CD44, CD24, CD133 and MIC-A/B (NKG2 receptors) in breast (MCF7) and ovarian (SK-OV-3) cells resistant to gemcitabine (GEM), paclitaxel (PTX) and 5-fluorouracil (5-FU) and (2) their elimination by Numb- and Notch-peptide activated CTL. The number of cells in all populations with the luminal CSC phenotype [epithelial specific antigen+ (ESA) CD44hi CD24lo, CD44hi CD133+, and CD133+ CD24lo] increased in drug-resistant MCF7 and SK-OV-3 cells. Similarly, the number of cells with expressed MIC-A/B increased 4 times in drug-resistant tumor cells compared with drug-sensitive cells. GEMRes MCF7 cells had lower levels of the Notch-1-extracellular domain (NECD) and Notch trans-membrane intracellular domain (TMIC) than GEMSens MCF7. The levels of Numb, and Numb-L-[P]-Ser265 were similar in GEMRes and GEMSens MCF7 cells. Only the levels of Numb-L (long)-Ser295 decreased slightly. This finding suggests that Notch-1 cleavage to TMIC is inhibited in GEMRes MCF7 cells. PBMC activated by natural immunogenic peptides Notch-1 (2112–2120) and Numb-1 (87–95) eliminated NICDpositive, CD24hi CD24lo MCF7 cells. It is likely that the immunogenic Numb-1 peptide in MCF7 cells originated from Numb, [P]-lated by an unknown kinase, because staurosporine but not wortmannin and MAPK-inhibitors decreased peptide presentation. Numb and Notch are antagonistic proteins which degrade each other to stop and activate cell proliferation, respectively. Their peptides are presented alternatively. Targeting both antagonistic proteins should be useful to prevent metastases in patients whose tumors are resistant to conventional treatments.

Electronic supplementary material

The online version of this article (doi:10.1007/s00262-008-0623-1) contains supplementary material, which is available to authorized users.

Keywords: Drug resistance, Breast/ovarian, Cancer stem cell, Notch, Numb, Peptide

Introduction

Renewal of embryonic (ESC) and adult stem cells (ASC) is regulated by signals from the surrounding environment. It is possible that cancer stem cells (CSC) originate from normal-ASC, or their intermediate progenitors (IP) which accumulate oncogenic mutations. More recently, they were shown to originate from ASC which activate embryonal differentiation programs. Regardless of their origin CSC renew after activation by Notch and amplification by Hedgehog and β-catenin (Wnt)-signals of Notch signals [2, 20, 23].

Breast cancer cells with high levels of CD44 (CD44hi) and absent or low CD24 (CD24neg/lo) expression have functional characteristics of CSC. Brain, colon and prostate cancer cells with CSC-characteristics express CD133 (prominin-1) [1, 3, 10, 28, 31].

The levels of CD44 (an adhesion molecule which binds to hyaluronate) directly correlate with metastasis in breast cancer. The levels of CD24 (an adhesion molecule that binds P-selectin) inversely correlate with survival of ovarian cancer patients. Studies in human mammary cells suggest that the most primitive mammary cells do not express estrogen-receptors, but are capable to differentiate to estrogen positive, luminal epithelial cells [6, 18, 24, 33].

CSC are more resistant to chemo- and radiotherapy than non-CSC [17, 25]. Histone-deacetylase (HDAC) and poly-A-ribose polymerase (PARP)-inhibitors are weak anti-CSC effectors. To avoid toxicity, it was proposed to lower doses of standard chemotherapy when used together with HDAC/PARP inhibitors [12, 26, 27].

A novel approach to cancer therapy is to eliminate CSC with immune effectors. We hypothesized that we can eliminate CSC by targeting peptides from the antagonistic Notch and Numb proteins with cytotoxic T lymphocytes (CTL). When CSC renew or become quiescent Notch and Numb induce degradation of each other, respectively. Notch and Numb are degraded to peptides by the proteasome. Peptides from NICD and Numb proteins are presented by HLA-class I molecules of cancer cells to T cells.

Hetero-dimeric Notch (hNotch) consists of an extracellular domain named NECD and a trans-membrane and intracellular domain, named NICD. NICD and NECD are linked by disulfide bonds. hNotch, located trans-membrane, is activated by its ligands. Human Notch-ligands belong to the Jagged (Jag) and Delta-like (DLL) families. Activation/inhibition of Notch can proceed in Cis- by Notch ligands expressed on the responding cell or in Trans-from Notch-ligand expressed by neighboring cells. Notch signals are transduced i by NICD, which activates gene transcription [14].

Numb has four isoforms. They form two groups Numb-Long (Numb-L) and Numb-Short (Numb-S) which differ in length by 5 kDa. Each group has two isoforms, which differ by 1 kDa, and are difficult to distinguish. Both Numb-L and Numb-S are present in cancer cells. However, most human cancer studies do not resolve Numb-S from Numb-L and Numb-like [19]. Non-phosphorylated Numb is adjacent to membrane. After receiving external signals Numb is [P]-lated at Ser295 and migrates to cytoplasm. In cytoplasm Numb-[P]-Ser295 cannot bind NICD to block cell-cycle activation. [P]-lation of Numb at other/additional sites directs its degradation by proteasome. The cells which degrade most Numb divide symmetrically, while the cells which do not degrade cytoplasmic Numb divide asymmetrically [4, 32].

Symmetric division of stem cells which lack Numb, double the number of “mother” stem cells. Asymmetric division of cells which contain both Notch and Numb results in one “mother” stem cell and one different cell [daughter/IP-stem cell [4, 32]]. Degradation of NICD by Numb inhibits activation of transcription by NICD and cells become quiescent [9, 11, 19].

The aim of this study was to determine whether: (1) the proportion of cells with CSC-phenotype/putative CSC increases in drug-resistant cancer lines, and (2) drug-resistant cells with CSC-phenotype can be eliminated by Notch- and Numb-peptide activated CTL.

We found higher numbers of CD44hi CD24lo, CD44hi CD133+ and CD24lo CD133+ cells in gemcitabine (GEMRes), paclitaxel (PTXRes) and 5-fluorouracil (5-FURes) resistant breast and ovarian cancer lines compared with Drug sensitive (DrugSens) cells. Three-four times more Drug resistant (DrugRes) cells expressed NKG2D receptors than DrugSens cells. NICD+ and CD44hi CD24lo cells were eliminated by Notch-1 (2112–2120) and Numb-1 (87–95) peptide-activated peripheral blood mononuclear cells (PBMC) and to a lesser extent by interleukin (IL)-2-activated PBMC.

Materials and methods

Cell lines and drugs

MCF7 and SK-OV-3 cell lines were from ATCC (Manassas, VA). Cells were cultured in RPMI 1640 medium with 10% FBS, 100 U/L penicillin, and 100 μg/mL streptomycin. We used gemcitabine (Eli-Lilly Indianapolis, IN), paclitaxel (Bristol-Myers Squibb, Princeton, NJ), 5-fluorourcil (Sigma Chemical Co. St Louis, MO) and human recombinant DLL4 (rhDLL4, R&D Systems, Inc. Minneapolis, MN).

Antibodies

The mAb to human antigens included anti-ESA (Biomeda, Foster City, CA), anti-CD44 (BD Pharmingen, San Diego, CA), anti-CD44 (BD), anti-CD24 (BD), anti-CD24 (Abcam Inc., Cambridge, MA), anti-MHC class I chain-related gene A (MICA)/MHC class I chain-related gene B (MICB) (R&D Systems, Minneapolis, MN), anti-CD133/2 (Miltenyi Biotec Inc., Auburn, CA), and anti-Notch-1 (BD). Polyclonal Abs included anti-Notch-1 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA), anti-NICD, anti-Numb, and anti-Bcl-2 mAb (Abcam).

mAb against rat Numb [P]-Ser264 and Numb [P]-Ser283 = Human Numb-L-P-Ser265 and 295 were prepared by Dr. Hiroshi Tokumitsu [34, 35]. Numb has many N-terminal splice variants. The position of the corresponding residues to [P]-Ser264 and [P]-Ser283 of rat Numb differs among isoforms. In human Numb-S is [P]-Ser265 and [P]-Ser284 [Numb-3 (NP_003735) and Numb-4 (NP_0010057450)]. Rat [P]-Ser276 is [P]-Ser295 in human Numb-L [Numb-1 (NP_001005743) and Numb-2 (NP_001005744)]. Rat [P]-Ser283 corresponds to Ser295 in human Numb-L. We used the designations [P]-Ser265 and [P]-Ser295 since they are close to the [P]-Ser positions reported in the recent literature.

IC50 of anticancer drugs

The 50% inhibitory concentration (IC50) of GEM, PTX and 5-FU was determined after a 72-h incubation with drugs as described [13].

Flow cytometry

Cells (25–30 × 106) were cultured with drugs at 2 × IC50 for 4 days followed by drug at 0.5–1.0 × IC50 for the next 3–6 days. Surviving cells (2 × 105) were incubated first with 20 μg of human IgG (Sigma) for 1 h on ice to inhibit nonspecific binding of specific mAb during staining. Analysis was performed using a Becton Dickinson FACSCalibur with CellQuest software (BD). Expression of CD24, CD44 and CD133 was quantified in gated ESA+ cells as described [16].

CSC-like and their progenitors were characterized following the geometrical mean (x 2) of CD24 in CD44hi cells. CD24negative = MFI, 0–10, CD24lo = MFI, 10–100, CD24hi = MFI, 100–1,000 [16]. ESA+ [(CD44hi and CD24lo/hi), and (CD133+)] cells were quantified using the formula: (% of total ESA+ cells) × (% of cells positive for both markers) × (number of cells in the culture).

Activation of GEMRes MCF7 cells

Cells were stimulated for 24 h in serum free medium with 62.5 ng/mL rhDLL4 then cultured with or without GEM for 7 days. GEMRes MCF7 cells were stimulated three times with DLL4 (62.5 ng/mL) weakly for 24 h.

Sensitivity of GEMRes MCF7 cells to HLA-A2+ PBMC activated by Notch- and Numb-peptides

Notch-1 = NICD peptide (2,112–2,120) and Numb-1 = Numb-1-PTB domain peptide (87–95) were previously identified by us as immunogenic [15]. Non-adherent PBMC were activated with peptide-pulsed autologous immature monocyte-derived dendritic cells (iDC), or T2 cells as we described [37]. In all activation procedures, we used IL-12 as co-factor followed by IL-2 48 h later.

Immunoselection

Because autologous T cells to MCF-7 are not available, we used allogeneic immunoselection. GEMRes MCF7 cells (1.0 × 105) were co-cultured in six-well plates with 3.0 × 105 IL-2-activated, Notch-1-, or Numb-1-peptide-activated PBMC for 5 days. IL-2 was not added to cultures for 72 h before use. Residual IL-2 was washed out. Surviving MCF7 cells were analyzed by flow-cytometry.

Western blot

Cell lysates with nuclei removed were prepared from live MCF7, and SK-OV-3 cells. Immunoblotting and quantification of NECD, NICD, Numb, and Bcl-2 utilizing β-actin as a reference was performed in relation to actin in the same sample, as we described [8, 37].

Results

The number of ESA+ [(CD44+ CD24lo), (CD44+ CD133+), and (CD24lo CD133+)] cells increase in drug-resistant MCF7 and SK-OV-3 cells

We quantified the IC50 of GEM, 5-FU and PTX for MCF7 and SKOV-3 cells. The IC50 (GEM) for MCF7 cells was 430 nM and the IC50 (GEM) for SK-OV-3 cells was 16.1 nM. The IC50 (5-FU) for MCF7 cells was 1,302 nM. The IC50 (5-FU) for SK-OV-3 cells was 3,560 nM. MCF7 cells were more sensitive to 5-FU than SK-OV-3 cells, and more resistant to GEM than SK-OV-3 cells. The IC50 (PTX) was similar for both cell lines = 4–5 nM.

To verify that ESA levels are the same in drug-resistant and sensitive cells we determined expression of ESA. The levels of ESA were the same in DrugRes and DrugSens cells (Fig. 1). DrugRes MCF7 cells were luminal cells (Fig. 1 and Supplementary Table 1). The number of cells with CSC-markers increased five and tenfold in MCF7 and SK-OV-3 cells, respectively. 5-FU and PTX selected CSC-like cells with similar efficiency with GEM. The number of CD133+, CD133+ CD44hi and CD133+ CD24lo cells increased by 3–5 fold in GEMRes MCF7 cells and GEMRes SK-OV-3 cells compared with the DrugSens cells (Fig. 2d).

Fig. 1.

Fig. 1

a, b Increase in number of CD44+ CD24lo cells in GEMRes MCF7 and SK-OV-3 cells. MCF7 [MFI (CD24lo)] cells range 10–100. c CD24lo MCF7 cells (MFI between 5 and 50) increase in number in 5-FURes and PTXRes MCF7 cells. This finding was confirmed in two additional independently performed experiments

Fig. 2.

Fig. 2

CD133+ cells increased in number in GEMRes cells. (*) significant increase >twofold. Untreated cells (black columns); GEMRes cells (white columns)

GEMRes MCF7 cells have lower levels of NECD than GEMSens cells

We found differences in the expression of antagonistic proteins Notch and Numb, between DrugRes and DrugSens cells. Reducing conditions break the disulfide bond between NECD and TMIC. TMIC is further cleaved between Ala1710 and Val1717 to generate the Notch-extracellular truncated domain (NEXT). NEXT is further cleaved at Val1744 to generate free NICD [5, 21, 30].

NECD levels decreased in GEMRes MCF7 cells compared with GEMSens MCF7 cells. Longer ER-precursors of Notch-1 were present in similar amounts in GEMRes and GEMSens MCF7 cells (Fig. 3a). TMIC levels also decreased in GEMRes MCF7 cells. The amount of NICD was low and similar in GEMRes and GEMSens cells. The decrease in Notch-1 suggested that Notch-1-NECD was degraded and not replaced, since the amount of its ER-precursor was higher. This means that less of Notch-1-ER-precursor was cleaved to TMIC (Fig. 3a). The levels of TMIC were lower than in control cells. The levels of Numb were slightly lower in GEMRes and untreated MCF7 cells (Fig. 3a). The ratio of NECD and TMIC (pre-NICD) to Numb-L and Numb-S decreased (see OD values). Therefore “quiescent” GEMRes MCF7 cells have lower levels of Notch-NECD than “quiescent” GEMSens MCF7 cells. Control, dividing SK-OV-3 cells synthesized high amounts of Notch and expressed both TMIC (of 100 kDa) and NICD (of 80 kDa) (Fig. 3c).

Fig. 3.

Fig. 3

a Decreased expression of Notch-1 (NECD) and Notch-1 (TMIC) in GEMRes MCF7 cells compared with untreated GEMSens MCF7 cells. One of the two experiments is shown b Numb-L and Numb-S are [P]-lated at Ser265 and Ser295 in GEMSens and GEMRes cells. MCF7 cells contain significantly less Numb-S than SKOV3 cells. c Numb is more [P]-lated at Ser265 than at Ser295 NECD was detected with mAbs-scc3275 (recognize the whole Notch molecule). H131 mAb detects two TMIC of 100 kDa and NICD of 80 kDa, respectively. One of two experiments is shown

Numb-L and Numb-S were [P]-lated at Ser295, in both untreated and GEMRes cells. The amount of Numb-[P]-Ser295 decreased by 30%, in GEMRes cells. The amount of Numb-L and Numb-S-[P]-Ser265 did not change in GEMRes cells compared with untreated cells (Fig. 3b). Our results indicate differences in Ser295-phosphorylation between GEMSens and GEMRes cells.

SK-OV-3 cells had more Numb-S than Numb-L. Furthermore SK-OV-3 cells had a lower ratio of Numb-L/S-[P]-Ser295 to Numb-L/S than MCF7 cells and ratios of Numb-L/S-[P]-Ser265 to Numb-L/S similar to those in MCF7 cells (Fig. 3c).

Soluble DLL4 expanded more CD24hi cells from GEMRes cells

The mRNA profile of Notch family members and of Notch ligands in MCF7 cells, untreated and treated with chemotherapeutics, has been reported. MCF7 has lower levels of Notch-ligands of the Delta-like family and higher levels of Notch-ligands of the Jagged-family [39]. MCF7 cells have similar levels of Notch-1 and Notch-2 with normal cells but significantly lower levels of Notch-3 and 4. We investigated the effects of Notch activation by its ligand, DLL4, on expansion of MCF7 cells. DLL4 expanded more CD24hi than CD24lo cells. DLL4 did not expand CD24hi cells in the presence of GEM (Supplementary Table 2). Many rhDLL4-expanded cells were CD44lo CD24lo and CD44lo CD24hi (not shown). Therefore, rhDLL4 did not preferentially stimulate proliferation of CD24neg/lo cells.

Naturally immunogenic Notch and Numb peptides expand Ag-specific CD8+ T cells

Our results show that a part of Numb-L/S is marked by [P]-lation. These findings suggest that cells with CSC-markers can be eliminated by HLA-A2-Notch-1, and Numb-1-peptide: complex-specific CTL.

Peptides Notch-1 and Numb-1 expanded Notch-1+-TCR+ CD8+ and Numb-1+-TCR+ CD8+ cells from the PBMC of a HLA-A2+ healthy donor (Fig. 4a, b). The Numb-1 peptide was more immunogenic than the Notch-1 peptide because it activated more CD8+ cells at 1 μM, whereas Notch-1 did so at a concentration of 5 μM.

Fig. 4.

Fig. 4

Peptides Notch-1 and Numb-1 expanded antigen-specific CD8+ cells. a Notch-1-TCR+ CD8+ cells expanded by incubation with 5 μg Notch-1 (2112–212)and b Numb-1-TCR+ CD8+ cells expanded by incubation with 1 μg Numb-1 (87–95). TCRhi, TCRmed, and TCRlo, populations are shown in gates R2, R3 and R4. Numbers in each box indicate the percentage of Ag-specific cells in the entire population. In the absence of Ag, IL-2 induced expansion of Ag-specific cells, was: Notch-1-TCR+ cells = (TCRhi: 0.1%, TCRmed: 0.1%, TCRlo: 1.0%). Numb-1-TCR+ cells = (TCRhi: 0.3%, TCRmed: 1.0%, and TCRlo: 0.4%) in PBMC from the same donor cultured with IL-2. c SK-OV-3.A2 cells present Numb-1peptide to Numb-1 peptide-activated PBMC. d Presentation of Numb-1 peptide to Numb-1 peptide-activated cells is dependent on [P]-lation by protein-Ser/Thr-kinases PI3 K does not appear to be involved in peptide presentation as shown by lack of effect of wortmannin. The MAPK-kinase inhibitor SB20380 had a weak inhibitory effect. IFN-γ was quantified at 24 h (Closed symbols) and at 48 h (Open symbols). Numb-1 peptide-activated PBMC produced more IFN-γ than Notch peptide-activated PBMC. At 48 h the amount of IFN- γ produced by two Notch peptide-activated cell lines was similar with the amount produced by the IL-2-activated cell lines. Only Notch-1 peptide can be presented by HLA-A2 antigens after Notch digestion by proteasome according to the program paproc.de

Numb-1 peptide-activated PBMC produced IFN-γ when incubated with SK-OV-3.A2 cells

Numb-1 and NICD-1 peptide-activated PBMC produced similar amounts of IFN-γ, in the first 24 h of co-culture with SK-OV-3.A2 cells. The same levels of IFN-γ were produced by control peptide, Notch-1-1947, which is not generated by proteasome. The SK-OV-3.A2 cell line acquires expression of HLA-A2 following transfection with a HLA-A2 expression plasmid. IFN-γ produced by Numb-1-activated cells doubled at 48 h of co-culture. The amount of IFN-γ produced by Notch-1-activated cells did not increase and remained similar to the amount produced by IL-2 activated cells (Fig. 4c). Therefore, either SK-OV-3 cells presented more Numb-1 peptide than Notch-1 peptide to CD8+ cells, or Numb-1-CD8+ cells have higher functional avidity for HLA-A2-Numb-1 peptide complexes.

To identify whether Numb-degradation is activated by [P]-lation, we repeated the experiment with inhibitors of Ser–Thr-kinases Wortmanin did not inhibit presentation of the Numb-1 peptide, while SB-20380 had a marginal late effect (Fig. 4d). The strongest inhibition of Numb-1 peptide presentation was mediated by staurosporine, a broad-spectrum inhibitor of protein–serine–threonine kinase family, indicating that an identified kinase is involved in Numb [P]-lation and degradation.

GEMRes MCF7 cells express more NKG2D ligands than GEMSens MCF7 cells

To determine whether cells with CSC-markers are sensitive to cellular effectors, other than Ag-specific CD8+ T cells, we quantified expression of MIC-A/-B in GEMRes, PTXRes and 5-FURes MCF7 cells. The percentage of MIC-A/B+ cells increased by 4.5 fold (83.9%) in CD44hi CD24lo GEMRes cells and by threefold (57.5%) in CD44hi CD24lo PTXRes MCF7 cells (Fig. 5a). The percentage of MIC-A/B+ CD133+ cells increased from 0.22 in GEMSens to 6.34 in GEMRes MCF7 cells (not shown). The mean fluorescence intensity values show that the density of MIC-A/B receptors per cell was similar in DrugSens and DrugRes MCF7 cells. Therefore, more drug-resistant CSC-like cells will be sensitive to NK/NK-T cells than DrugSens cells. However, the sensitivity of each CSC-like cell to NK/NK-T cells is not expected to increase compared with DrugSens cells.

Fig. 5.

Fig. 5

a The number of MIC-A/-B+ cells increased in drug-resistant MCF7. White peaks represent ESA+ cells. Black peaks represent the MIC-A/B+ CD44+ CD24lo cells. be Co-culture of GEMRes MCF7 cells with Notch-1 peptide-activated PBMC decrease the NICD-Notch+ cell numbers. Surviving NICD+ MCF7 cells after co-culture with: b no effectors. c IL-2 activated PMBC. d Notch-1 peptide-activated PBMC. e Numb-1 peptide-activated PMBC. The % of NICD+ cells is shown in the upper right quadrant. The decrease in NICD+ cells in relation to the NICD+ cells in panel (b) was: IL-2 activated PBMC, 4.4%; Notch-1-activated PBMC, 68.5%; and Numb-1-activated PBMC, 25.3%. Note: the amount of free NICD in GEMRes MCF7 cells was lower than in UT-MCF7 cells (Fig. 3). f Numb-1 peptide-activated PBMC eliminate CD44+ cells from UT-MCF7 cells. 50,000 GEMRes MCF7 cells were cultured with indicated effectors for 5 days. Then live cells were collected and analyzed for expression of CD44 and CD24 in the same experiment. Analysis was performed in large tumor cells of similar cellularity (FS: 800–1000, side scatter 100–800). All indicate all live cells regardless of phenotype, CD44hiCD24lo indicate CSC-like cells, CD44hiCD24hi indicate potentially metastatic MCF7 cells, CD44loCD24lo indicate non-invasive MCF7 cells. Open columns no effectors, NP, dotted columns MCF7 cultured with IL-2- activated PBMC, NO, dashed columns left to right MCF7 cultured with Notch-1-activated PBMC, NU, textured columns MCF7 cultured with Numb-1- activated PBMC

Allogeneic Notch and Numb peptide-activated PBMC eliminated cells with CSC-phenotype markers

We investigated whether IL-2-activated, Notch-activated, and Numb-activated allogenic PBMC eliminate cells with CSC markers. To account for elimination of cells with CSC markers by allogeneic effectors we repeated the experiments, in the presence of IL-2-activated PBMC, and quantified each surviving population of CD44 CD24 cells. Therefore, in addition to allo-recognition of tumor cells by effectors, a significant recognition was due to Numb-1 peptide activated T cells. Forty-five percent of GEMRes cells had detectable NICD (Fig. 5b). Notch-1-positive cells decreased by 68.5% (from 45.4 to 14.3%) after co-culture with Notch-1 peptide-activated PBMC (Fig. 5d). Numb-1 peptide-activated PBMC decreased the NICD+ cells only by 25.3% (from 45.4 to 33.9%), whereas IL-2-activated non-specific PBMC had no significant effect (Fig. 5c, e). Therefore, Notch-1-specific CD8+ T cells specifically eliminated NICD+ cells.

To eliminate cells with CSC markers we repeated the experiments and quantified each surviving population of CD44 CD24 cells. To increase stringency of elimination, we used as target MCF7 cells because they expressed less Numb-L and ten times less Numb-S than SK-OV-3 cells.

Numb-1-activated T cells were more effective than Notch-1 and IL-2-activated effectors. Only 10% of the initially plated GEMRes cells survived. The majority of surviving cells were CD44hi CD24lo. Numb-1-activated cells eliminated 2–3 times more CD44hi CD24lo MCF7 cells than other effectors (Fig. 5f. Columns All and CD44hi CD24lo). Per effecter-cell number, Numb-1-peptide activated cells were more effective than IL-2-activated cells, in eliminating CD44hi CD24hi cells (Fig. 5f, Column CD44hi CD24hi).

Discussion

We identified populations in breast and ovarian tumor cell lines which express the phenotypes of breast (ESA+/CD44+/CD24−/lo) and brain, colon and prostate (CD133+) CSC. Because their function was not yet characterized, we designated these populations as CSC-like. CSC-like cells increased in DrugRes MCF7 and SK-OV-3 cells. Our novel findings are: (1) CD24lo population increased in DrugRes cells regardless of the mechanism of drug action; (2) The brain, colon and prostate CSC marker, CD133, is present on cells which express the breast CSC-markers; (3) Cells with CSC-markers/CSC-like cells in GEMRes MCF7 cells were eliminated by Numb-1 peptide-activated PBMC.

GEM and 5-FU are inhibitors of DNA and RNA synthesis, which are incorporated in newly synthesized strands. Neither GEM nor 5-FU affects cells in G1 phase [36]. These cells survive because their nucleic acid synthesis is minimal. In contrast, PTX acts by epigenetic mechanisms, by interfering with polymerization of actin.

We found only small differences in proliferation of CD24 and CD24+ cells when stimulated with medium, and found differences when cells were stimulated with DLL4. Cell-cycle analysis indicates that most cells were in G1 phase and only a small fraction less than 5% were in G2 M phase (Supplemental Figure 1). We used stringent conditions for isolation of these cells by maintaining the drug in culture for several weeks. All non-adherent cells died in these conditions. In clinical practice, the drug given to patient starts decaying and only a small amount is present several days later. When MCF7 cells were cultured in these conditions we counted that only 5–7 clones out of 50,000 drug-treated cells survived for one month and proliferated.

Therefore, we could not establish whether the cells which we analyzed are bona fide CSC, IP or drug-resistant tumor cells, which shared the phenotype of CSC.

DrugRes cells activated autonomous proliferation after drugs were removed. The Notch ligand, DLL4, expanded better CD24hi cells than CD24lo cells. Notch-ligands on neighboring cells activate Notch in Trans; by endocytosing NECD from responder cells. Free and plastic bound Notch-ligands are weak activators of Notch. Free Notch ligands, can stimulate or inhibit cells in Cis [23]. DLL4 delayed proliferation of CD44lo cells suggesting that it was inhibitory as described in other systems [14, 21].

We found that the balance between Notch and its antagonist, Numb, in DrugRes cancer cells, shifted in favor of Numb. The amount of NECD and TMIC-NICD decreased significantly. The amount of Numb did not change. Numb was present in four forms: non-phosporylated, [P]-lated at Ser265, [P]-lated at Ser295 and probably phosphorylated at both positions. Numb-[P]-Ser295 decreased. Since Numb-1 presentation required [P]-lation it is likely that the Numb-1 peptide derived from Numb-[P]-Ser295.

Numb interacts with the aPKC binding partner, PAR-3. Phosphorylation of Numb may inhibit cell migration by inhibiting integrin endocytosis [35]. [P]-Numb does not bind integrins when [P]-lated by atypical protein kinase-C (aPKC). Numb-[P]-Ser295 recruits 14-3-3 proteins, which inhibit the binding with AP-2 complex in vitro [22]. Although the 14-3-3-ζ vector rescued Numb-S from degradation in normal MCF-10A cells, Numb-S was not protected by 14-3-3-ζ from degradation in MCF7 cells (Fig. 6).

Fig. 6.

Fig. 6

Protein expression of Notch and Numb in MCF10A and MCF7. a Expression of Notch-1-NECD was not different between the normal breast cell line MCF10A and breast cancer cell line MCF7, but MCF7 cells expressed Notch-ER-precursor stronger than MCF10A cells. b The 14-3-3-ζ vector transfected MCF10A cells protected degradation of Numb-S but the 14-3-3-ζ transfected MCF7 cells did not. The band of 14-3-3-ζ HA-tag indicates expression of transfected 14-3-3-ζ protein. Cells which were transfected vector control have the band of 14-3-3-ζ protein by nature but not the band of 14-3-3-ζ HA tag

In normal cells, the Polo-kinase-1, Plk-1 regulates Numb asymmetry by [P]-lation of Pon [38]. The use of Polo- and Aurora-kinase inhibitors, in cancer treatment, raises the question whether more CSC will remain temporarily quiescent, or they will activate Notch when Numb decays and accelerate cancer progression. If this is the case, Numb-1 CTL can eliminate Plk-1-inhibitor-surviving cells. When this paper was prepared for publication two independent studies estimated the number of cells with CSC-characteristics in MCF7 to 2.4%. MCF7 cells formed mammospheres and required IL-6 to upregulate Notch-3 [7, 29].

In conclusion, we found that Numb-1 specific CTL can eliminate CD44hi CD24neg/lo cells. Based on our findings, adoptive and active immunotherapy (vaccines) with Notch and Numb should be effective in eliminating “quiescent CSC” in patients with breast and ovarian cancer.

Electronic supplementary material

Below is the link to the electronic supplementary material.

References

  • 1.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100:3983–3988. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Androutsellis-Theotokis A, Leker RR, Soldner F, Hoeppner DJ, Ravin R, Poser SW, Rueger MA, Bae SK, Kittappa R, McKay RD. Notch signaling regulates stem cell numbers in vitro and in vivo. Nature. 2006;442:823–826. doi: 10.1038/nature04940. [DOI] [PubMed] [Google Scholar]
  • 3.Balic M, Lin H, Young L, Hawes D, Giuliano A, McNamara G, Datar RH, Cote RJ. Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin Cancer Res. 2006;12:5615–5621. doi: 10.1158/1078-0432.CCR-06-0169. [DOI] [PubMed] [Google Scholar]
  • 4.Berdnik D, Torok T, Gonzalez-Gaitan M, Knoblich JA. The endocytic protein a-adaptin is required for numb-mediated asymmetric cell division in Drosophila . Dev Cell. 2002;3:221–231. doi: 10.1016/S1534-5807(02)00215-0. [DOI] [PubMed] [Google Scholar]
  • 5.Blaumueller CM, Qi H, Zagouras P, Artavanis-Tsakonas S. Intracellular cleavage of Notch leads to a heterodimeric receptor on the plasma membrane. Cell. 1997;90(2):281–291. doi: 10.1016/S0092-8674(00)80336-0. [DOI] [PubMed] [Google Scholar]
  • 6.Bourguignon LY, Gilad E, Peyrollier K. Heregulin-mediated ErbB2-ERK signaling activates hyaluronan synthases leading to CD44-dependent ovarian tumor cell growth and migration. J Biol Chem. 2007;282:19426–19441. doi: 10.1074/jbc.M610054200. [DOI] [PubMed] [Google Scholar]
  • 7.Cariati M, Naderi A, Brown JP, Smalley MJ, Pinder SE, Caldas C, Purushotham AD. ã6 integrin is necessary for the tumourigenicity of a stem cell-like subpopulation within the MCF7 breast cancer cell line. Int J Cancer. 2008;122(2):298–304. doi: 10.1002/ijc.23103. [DOI] [PubMed] [Google Scholar]
  • 8.Castilleja A, Carter D, Efferson CL, Ward NE, Kawano K, Fisk B, Kudelka AP, Gershenson DM, Murray JL, O’Brian CA, Ioannides CG. Induction of tumor-reactive CTL by C-side chain variants of the CTL epitope HER-2/neu proto-oncogene (369–377) selected by molecular modeling of the peptide: HLA-A2 complex. J Immunol. 2002;169:3545–3554. doi: 10.4049/jimmunol.169.7.3545. [DOI] [PubMed] [Google Scholar]
  • 9.Chapman G, Liu L, Sahlgren C, Dahlqvist C, Lendahl U. High levels of Notch signaling down-regulate Numb and Numblike. J Cell Biol. 2006;175:535–540. doi: 10.1083/jcb.200602009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 2005;65:10946–10951. doi: 10.1158/0008-5472.CAN-05-2018. [DOI] [PubMed] [Google Scholar]
  • 11.Di Marcotullio L, Ferretti E, Greco A, De Smaele E, Po A, Sico MA, Alimandi M, Giannini G, Maroder M, Screpanti I, Gulino A. Numb is a suppressor of Hedgehog signalling and targets Gli1 for Itch-dependent ubiquitination. Nat Cell Biol. 2006;8:1415–1423. doi: 10.1038/ncb1510. [DOI] [PubMed] [Google Scholar]
  • 12.Edwards SL, Brough R, Lord CJ, Natrajan R, Vatcheva R, Levine DA, Boyd J, Reis-Filho JS, Ashworth A resistance to therapy caused by intragenic deletion in BRCA2. Nature. 2008;451:1111–1115. doi: 10.1038/nature06548. [DOI] [PubMed] [Google Scholar]
  • 13.Efferson CL, Tsuda N, Kawano K, Nistal-Villan E, Sellappan S, Yu D, Murray JL, Garcia-Sastre A, Ioannides CG. Prostate tumor cells infected with a recombinant influenza virus expressing a truncated NS1 protein activate cytolytic CD8 + cells to recognize noninfected tumor cells. J Virol. 2006;80:383–394. doi: 10.1128/JVI.80.1.383-394.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hurlbut GD, Kankel MW, Lake RJ, Artavanis-Tsakonas S. Crossing paths with Notch in the hyper-network. Curr Opin Cell Biol. 2007;2:166–175. doi: 10.1016/j.ceb.2007.02.012. [DOI] [PubMed] [Google Scholar]
  • 15.Ishiyama S, Matsueda S, Jones LA, Efferson C, Celestino J, Schmandt R, Ioannides CG, Tsuda N, Chang DZ. Novel natural immunogenic peptides from Numb1 and Notch1 proteins for CD8+ cells in ovarian ascites. Int J Oncol. 2007;30:889–898. [PubMed] [Google Scholar]
  • 16.Kawano K, Efferson CL, Peoples GE, Carter D, Tsuda N, Murray JL, Ioannides CG. Sensitivity of undifferentiated, high-TCR density CD8 + cells to methylene groups appended to tumor antigen determines their differentiation or death. Cancer Res. 2005;65:2930–2937. doi: 10.1158/0008-5472.CAN-04-2232. [DOI] [PubMed] [Google Scholar]
  • 17.Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, Hilsenbeck SG, Pavlick A, Zhang X, Chamness GC, Wong H, Rosen J, Chang JC. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst. 2008;100(9):672–679. doi: 10.1093/jnci/djn123. [DOI] [PubMed] [Google Scholar]
  • 18.Liu S, Ginestier C, Charafe-Jauffret E, Foco H, Kleer CG, Merajver SD, Dontu G, Wicha MS. BRCA1 regulates human mammary stem/progenitor cell fate. Proc Natl Acad Sci USA. 2008;105(5):1680–1685. doi: 10.1073/pnas.0711613105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.McGill MA, McGlade CJ. Mammalian Numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain. J Biol Chem. 2003;278:23196–23203. doi: 10.1074/jbc.M302827200. [DOI] [PubMed] [Google Scholar]
  • 20.Morrison SJ, Kimble J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature. 2006;441:1068–1074. doi: 10.1038/nature04956. [DOI] [PubMed] [Google Scholar]
  • 21.Mumm JS, Schroeter EH, Saxena MT, Griesemer A, Tian X, Pan DJ, Ray WJ, Kopan RA. Ligand-induced extracellular cleavage regulates gamma-secretase-like proteolytic activation of Notch1. Mol Cell. 2000;5(2):197–206. doi: 10.1016/S1097-2765(00)80416-5. [DOI] [PubMed] [Google Scholar]
  • 22.Nishimura T, Kaibuchi K. Numb controls integrin endocytosis for directional cell migration with aPKC and PAR-3. Dev Cell. 2007;1:15–28. doi: 10.1016/j.devcel.2007.05.003. [DOI] [PubMed] [Google Scholar]
  • 23.Orford KW, Scadden DT. Deconstructing stem cell self-renewal: genetic insights into cell-cycle regulation. Nat Rev Genet. 2008;9:115–128. doi: 10.1038/nrg2269. [DOI] [PubMed] [Google Scholar]
  • 24.Ouhtit A, Abd Elmageed ZY, Abdraboh ME, Lioe TF, Raj MH. In vivo evidence for the role of CD44 in promoting breast cancer metastasis to the liver. Am J Pathol. 2007;171(6):2033–2039. doi: 10.2353/ajpath.2007.070535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Phillips TM, McBride WH, Pajonk F. The response of CD24(-/low)/CD44 + breast cancer-initiating cells to radiation. J Natl Cancer Inst. 2006;98(24):1777–1785. doi: 10.1093/jnci/djj495. [DOI] [PubMed] [Google Scholar]
  • 26.Piekarz RL, Sackett DL, Bates SE. Histone deacetylase inhibitors and demethylating agents: clinical development of histone deacetylase inhibitors for cancer therapy. Cancer J. 2007;13(1):30–39. doi: 10.1097/PPO.0b013e31803c73cc. [DOI] [PubMed] [Google Scholar]
  • 27.Ratnam K, Low JA. Current development of clinical inhibitors of poly (ADP-ribose)-polymerase in oncology. Clin Cancer Res. 2007;13:1383–1388. doi: 10.1158/1078-0432.CCR-06-2260. [DOI] [PubMed] [Google Scholar]
  • 28.Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111–115. doi: 10.1038/nature05384. [DOI] [PubMed] [Google Scholar]
  • 29.Sansone P, Storci G, Tavolari S, Guarnieri T, Giovannini C, Taffurelli M, Ceccarelli C, Santini D, Paterini P, Marcu KB, Chieco P, Bonafè M. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest. 2007;117:3988–4002. doi: 10.1172/JCI32533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schroeter EH, Kisslinger JA, Kopan R. Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature. 1998;393(6683):382–386. doi: 10.1038/30756. [DOI] [PubMed] [Google Scholar]
  • 31.Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–5828. [PubMed] [Google Scholar]
  • 32.Smith CA, Lau KM, Rahmani Z, Dho SE, Brothers G, She YM, Berry DM, Bonneil E, Thibault P, Schweisguth F, Le Borgne R, McGlade CJ. aPKC-mediated phosphorylation regulates asymmetric membrane localization of the cell fate determinant Numb. EMBO J. 2007;26:468–480. doi: 10.1038/sj.emboj.7601495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Surowiak P, Materna V, Kaplenko I, Spaczyński M, Dietel M, Kristiansen G, Lage H, Zabel M. Unfavorable prognostic value of CD24 expression in sections from primary and relapsed ovarian cancer tissue. Int J Gynecol Cancer. 2006;2:515–521. doi: 10.1111/j.1525-1438.2006.00555.x. [DOI] [PubMed] [Google Scholar]
  • 34.Tokumitsu H, Hatano N, Inuzuka H, Sueyoshi Y, Yokokura S, Ichimura T, Nozaki N, Kobayashi R. Phosphorylation of Numb family proteins. Possible involvement of Ca2 +/calmodulin-dependent protein kinases. J Biol Chem. 2005;280:35108–35118. doi: 10.1074/jbc.M503912200. [DOI] [PubMed] [Google Scholar]
  • 35.Tokumitsu H, Hatano N, Yokokura S, Sueyoshi Y, Nozaki N, Kobayashi R. Phosphorylation of Numb regulates its interaction with the clathrin-associated adaptor AP-2. FEBS Lett. 2006;580(24):5797–5801. doi: 10.1016/j.febslet.2006.09.043. [DOI] [PubMed] [Google Scholar]
  • 36.Tolis C, Peters GJ, Ferreira CG, Pinedo HM, Giaccone G. Cell cycle disturbances and apoptosis induced by topotecan and gemcitabine on human lung cancer cell lines. Eur J Cancer. 1999;35:796–807. doi: 10.1016/S0959-8049(98)00425-0. [DOI] [PubMed] [Google Scholar]
  • 37.Tsuda N, Chang DZ, Mine T, Efferson C, García-Sastre A, Wang X, Ferrone S, Ioannides CG. Taxol increases the amount and T cell activating ability of self-immune stimulatory multimolecular complexes found in ovarian cancer cells. Cancer Res. 2007;67:8378–8387. doi: 10.1158/0008-5472.CAN-07-0327. [DOI] [PubMed] [Google Scholar]
  • 38.Wang H, Ouyang Y, Somers WG, Chia W, Lu B. Polo inhibits progenitor self-renewal and regulates Numb asymmetry by phosphorylating Pon. Nature. 2007;449(7158):96–100. doi: 10.1038/nature06056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.http://www.ncbi.nlm.nih.gov/geo/gds/profileGraph

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES